ijms-logo

Journal Browser

Journal Browser

TFF Peptides: Lectins in Mucosal Protection and More

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Biochemistry".

Deadline for manuscript submissions: closed (8 October 2019) | Viewed by 55353

Special Issue Editors


E-Mail Website
Guest Editor
Director of the Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
Interests: protection, regeneration and repair of mucous epithelia with special emphasis on the role of TFF peptides; TFF peptides in the immune and central nervous systems
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Head of the Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsstr. 19, 91054 Erlangen, Germany
Interests: protein and peptide research (mucins, TFF peptides, surfactant proteins, antimicrobial peptides, innate immune system, ocular surface, and lacrimal apparatus)

Special Issue Information

Dear Colleagues,

 

Trefoil factor family (TFF) peptides—together with mucins—are characteristic secretory products of mucous epithelia, where they are involved in different protective mechanisms. For example, they show motogenic and (anti)apoptotic activities. Furthermore, TFFs are also expressed in both the central nervous and immune systems. Pathologically, TFFs are ectopically expressed in various tumors, in inflammatory diseases, and after wounding. Transgenic TFF-deficient mice show different abnormalities in the gastrointestinal tract and the immune system, respectively. Based on these results, TFFs have considerable therapeutic potential, e.g., for treating certain inflammatory disorders as well as oral mucositis after radio- and chemotherapy. Thus far, no high-affinity binding of a TFF peptide with a classical transmembrane receptor has been identified. However, there are numerous interactions of TFFs with (glyco)proteins and a gastrokine documented. The specific interaction particularly of TFF1 and TFF2 with carbohydrate moieties clearly established them as lectins.

Papers submitted to this Special Issue must report high novelty results, e.g., concerning the complex biosynthesis of TFFs, the characterization of TFF interaction partners, the molecular function of TFFs, and the characterization of TFF modules in mosaic proteins. Additionally, state-of-the-art reviews concerning selected functional aspects of TFFs are welcome.

 

Prof. Dr. Werner Hoffmann
Prof. Dr. Friedrich Paulsen
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Apoptosis
  • Cell migration
  • Epithelia
  • Inflammation
  • Lectin
  • Mucin
  • Mucosal protection
  • Neuropeptide
  • TFF domain
  • Trefoil.

Published Papers (16 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

18 pages, 3419 KiB  
Article
The Tumor Suppressor TFF1 Occurs in Different Forms and Interacts with Multiple Partners in the Human Gastric Mucus Barrier: Indications for Diverse Protective Functions
by Jörn Heuer, Franziska Heuer, René Stürmer, Sönke Harder, Hartmut Schlüter, Nayara Braga Emidio, Markus Muttenthaler, Dörthe Jechorek, Frank Meyer and Werner Hoffmann
Int. J. Mol. Sci. 2020, 21(7), 2508; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21072508 - 04 Apr 2020
Cited by 24 | Viewed by 3220
Abstract
TFF1 is a protective peptide of the Trefoil Factor Family (TFF), which is co-secreted with the mucin MUC5AC, gastrokine 2 (GKN2), and IgG Fc binding protein (FCGBP) from gastric surface mucous cells. Tff1-deficient mice obligatorily develop antropyloric adenoma and about 30% progress [...] Read more.
TFF1 is a protective peptide of the Trefoil Factor Family (TFF), which is co-secreted with the mucin MUC5AC, gastrokine 2 (GKN2), and IgG Fc binding protein (FCGBP) from gastric surface mucous cells. Tff1-deficient mice obligatorily develop antropyloric adenoma and about 30% progress to carcinomas, indicating that Tff1 is a tumor suppressor. As a hallmark, TFF1 contains seven cysteine residues with three disulfide bonds stabilizing the conserved TFF domain. Here, we systematically investigated the molecular forms of TFF1 in the human gastric mucosa. TFF1 mainly occurs in an unusual monomeric form, but also as a homodimer. Furthermore, minor amounts of TFF1 form heterodimers with GKN2, FCGBP, and an unknown partner protein, respectively. TFF1 also binds to the mucin MUC6 in vitro, as shown by overlay assays with synthetic 125I-labeled TFF1 homodimer. The dominant presence of a monomeric form with a free thiol group at Cys-58 is in agreement with previous studies in Xenopus laevis and mouse. Cys-58 is likely highly reactive due to flanking acid residues (PPEEEC58EF) and might act as a scavenger for extracellular reactive oxygen/nitrogen species protecting the gastric mucosa from damage by oxidative stress, e.g., H2O2 generated by dual oxidase (DUOX). Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Graphical abstract

11 pages, 2446 KiB  
Article
Trefoil Factor Family (TFF) Modules Are Characteristic Constituents of Separate Mucin Complexes in the Xenopus laevis Integumentary Mucus: In Vitro Binding Studies with FIM-A.1
by René Stürmer, Jana Reising and Werner Hoffmann
Int. J. Mol. Sci. 2020, 21(7), 2400; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21072400 - 31 Mar 2020
Cited by 4 | Viewed by 2541
Abstract
The skin of the frog Xenopus laeevis is protected from microbial infections by a mucus barrier that contains frog integumentary mucins (FIM)-A.1, FIM-B.1, and FIM-C.1. These gel-forming mucins are synthesized in mucous glands consisting of ordinary mucous cells and one or more cone [...] Read more.
The skin of the frog Xenopus laeevis is protected from microbial infections by a mucus barrier that contains frog integumentary mucins (FIM)-A.1, FIM-B.1, and FIM-C.1. These gel-forming mucins are synthesized in mucous glands consisting of ordinary mucous cells and one or more cone cells at the gland base. FIM-A.1 and FIM-C.1 are unique because their cysteine-rich domains belong to the trefoil factor family (TFF). Furthermore, FIM-A.1 is unusually short (about 400 amino acid residues). In contrast, FIM-B.1 contains cysteine-rich von Willebrand D (vWD) domains. Here, we separate skin extracts by the use of size exclusion chromatography and analyze the distribution of FIM-A.1 and FIM-C.1. Two mucin complexes were detected, i.e., a high-molecular-mass Complex I, which contains FIM-C.1 and little FIM-A.1, whereas Complex II is of lower molecular mass and contains the bulk of FIM-A.1. We purified FIM-A.1 by a combination of size-exclusion chromatography (SEC) and anion-exchange chromatography and performed first in vitro binding studies with radioactively labeled FIM-A.1. Binding of 125I-labeled FIM-A.1 to the high-molecular-mass Complex I was observed. We hypothesize that the presence of FIM-A.1 in Complex I is likely due to lectin interactions, e.g., with FIM-C.1, creating a complex mucus network. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Graphical abstract

9 pages, 2274 KiB  
Article
Subcellular Localization of the TFF Peptides xP1 and xP4 in the Xenopus laevis Gastric/Esophageal Mucosa: Different Secretion Modes Reflecting Diverse Protective Functions
by Heinz Schwarz and Werner Hoffmann
Int. J. Mol. Sci. 2020, 21(3), 761; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21030761 - 23 Jan 2020
Cited by 7 | Viewed by 2347
Abstract
The TFF peptides xP1 and xP4 from Xenopus laevis are orthologs of TFF1 and TFF2, respectively. xP1 is secreted as a monomer from gastric surface mucous cells and is generally not associated with mucins, whereas xP4 is a typical secretory peptide from esophageal [...] Read more.
The TFF peptides xP1 and xP4 from Xenopus laevis are orthologs of TFF1 and TFF2, respectively. xP1 is secreted as a monomer from gastric surface mucous cells and is generally not associated with mucins, whereas xP4 is a typical secretory peptide from esophageal goblet cells, and gastric mucous neck and antral gland cells tightly associated as a lectin with the ortholog of mucin MUC6. Both TFF peptides have diverse protective functions, xP1 as a scavenger for reactive oxygen species preventing oxidative damage and xP4 as a constituent of the water-insoluble adherent inner mucus barrier. Here, we present localization studies using immunofluorescence and immunoelectron microscopy. xP1 is concentrated in dense cores of secretory granules of surface mucous cells, whereas xP4 mixes with MUC6 in esophageal goblet cells. Of note, we observe two different types of goblet cells, which differ in their xP4 synthesis, and this is even visible morphologically at the electron microscopic level. xP4-negative granules are recognized by their halo, which is probably the result of shrinkage during the processing of samples for electron microscopy. Probably, the tight lectin binding of xP4 and MUC6 creates a crosslinked mucous network forming a stabile granule matrix, which prevents shrinkage. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Graphical abstract

16 pages, 1964 KiB  
Article
Molecular Alterations in the Stomach of Tff1-Deficient Mice: Early Steps in Antral Carcinogenesis
by Eva B. Znalesniak, Franz Salm and Werner Hoffmann
Int. J. Mol. Sci. 2020, 21(2), 644; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21020644 - 18 Jan 2020
Cited by 17 | Viewed by 3506
Abstract
TFF1 is a peptide of the gastric mucosa co-secreted with the mucin MUC5AC. It plays a key role in gastric mucosal protection and repair. Tff1-deficient (Tff1KO) mice obligatorily develop antropyloric adenoma and about 30% progress to carcinomas. Thus, these [...] Read more.
TFF1 is a peptide of the gastric mucosa co-secreted with the mucin MUC5AC. It plays a key role in gastric mucosal protection and repair. Tff1-deficient (Tff1KO) mice obligatorily develop antropyloric adenoma and about 30% progress to carcinomas. Thus, these mice represent a model for gastric tumorigenesis. Here, we compared the expression of selected genes in Tff1KO mice and the corresponding wild-type animals (RT-PCR analyses). Furthermore, we systematically investigated the different molecular forms of Tff1 and its heterodimer partner gastrokine-2 (Gkn2) in the stomach (Western blot analyses). As a hallmark, a large portion of murine Tff1 occurs in a monomeric form. This is unexpected because of its odd number of seven cysteine residues. Probably the three conserved acid amino acid residues (EEE) flanking the 7th cysteine residue allow monomeric secretion. As a consequence, the free thiol of monomeric Tff1 could have a protective scavenger function, e.g., for reactive oxygen/nitrogen species. Furthermore, a minor subset of Tff1 forms a disulfide-linked heterodimer with IgG Fc binding protein (Fcgbp). Of special note, in Tff1KO animals a homodimeric form of Gkn2 was observed. In addition, Tff1KO animals showed strongly reduced Tff2 transcript and protein levels, which might explain their increased sensitivity to Helicobacter pylori infection. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Figure 1

21 pages, 6690 KiB  
Article
Pharmacological Inhibition of TFF3 Enhances Sensitivity of CMS4 Colorectal Carcinoma to 5-Fluorouracil through Inhibition of p44/42 MAPK
by Ru-Mei Chen, Yi-Shiou Chiou, Qing-Yun Chong, Han-Ming Poh, Tuan-Zea Tan, Meng-Yi Zhang, Lan Ma, Tao Zhu, Vijay Pandey, Basappa, Alan Prem Kumar and Peter E. Lobie
Int. J. Mol. Sci. 2019, 20(24), 6215; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20246215 - 09 Dec 2019
Cited by 16 | Viewed by 3729
Abstract
Increased expression of trefoil factor 3 (TFF3) has been reported in colorectal carcinoma (CRC), being correlated with distant metastasis and poor clinical outcomes. Amongst the CRC subtypes, mesenchymal (CMS4) CRC is associated with the worst survival outcome. Herein, the functional roles of TFF3 [...] Read more.
Increased expression of trefoil factor 3 (TFF3) has been reported in colorectal carcinoma (CRC), being correlated with distant metastasis and poor clinical outcomes. Amongst the CRC subtypes, mesenchymal (CMS4) CRC is associated with the worst survival outcome. Herein, the functional roles of TFF3 and the pharmacological inhibition of TFF3 by a novel specific small molecule TFF3 inhibitor—2-amino-4-(4-(6-fluoro-5-methylpyridin-3-yl)phenyl)-5-oxo-4H,5H-pyrano[3,2-c]chromene-3-carbonitrile (AMPC) in CMS4 CRC was explored. Forced expression of TFF3 in CMS4 CRC cells promoted cell proliferation, cell survival, foci formation, invasion, migration, cancer stem cell like behaviour and growth in 3D Matrigel. In contrast, siRNA-mediated depletion of TFF3 or AMPC inhibition of TFF3 in CMS4 CRC cells decreased oncogenic behaviour as indicated by the above cell function assays. AMPC also inhibited tumour growth in vivo. The TFF3-stimulated oncogenic behaviour of CMS4 CRC cells was dependent on TFF3 activation of the p44/42 MAPK (ERK1/2) pathway. Furthermore, the forced expression of TFF3 decreased the sensitivity of CMS4 CRC cells to 5-fluorouracil (5-FU); while depleted TFF3 expression enhanced 5-FU sensitivity in CMS4 CRC cells. 5-FU treatment induced TFF3 expression in CMS4 CRC cells. AMPC, when used in combination with 5-FU in CMS4 CRC cells exhibited a synergistic inhibitory effect. In summary, this study provides functional evidence for TFF3 as a therapeutic target in CMS4 CRC. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Figure 1

15 pages, 1302 KiB  
Article
Human Synovia Contains Trefoil Factor Family (TFF) Peptides 1–3 Although Synovial Membrane Only Produces TFF3: Implications in Osteoarthritis and Rheumatoid Arthritis
by Judith Popp, Martin Schicht, Fabian Garreis, Patricia Klinger, Kolja Gelse, Stefan Sesselmann, Michael Tsokos, Saskia Etzold, Dankwart Stiller, Horst Claassen and Friedrich Paulsen
Int. J. Mol. Sci. 2019, 20(23), 6105; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20236105 - 03 Dec 2019
Cited by 4 | Viewed by 3705
Abstract
Objective: Trefoil factor family peptide 3 (TFF3) has been shown to support catabolic functions in cases of osteoarthritis (OA). As in joint physiology and diseases such as OA, the synovial membrane (SM) of the joint capsule also plays a central role. We analyze [...] Read more.
Objective: Trefoil factor family peptide 3 (TFF3) has been shown to support catabolic functions in cases of osteoarthritis (OA). As in joint physiology and diseases such as OA, the synovial membrane (SM) of the joint capsule also plays a central role. We analyze the ability of SM to produce TFF compare healthy SM and its secretion product synovial fluid (SF) with SM and SF from patients suffering from OA or rheumatoid arthritis (RA). Methods: Real-time PCR and ELISA were used to measure the expression of TFFs in healthy SM and SM from patients suffering from OA or RA. For tissue localization, we investigated TFF1-3 in differently aged human SM of healthy donors by means of immunohistochemistry, real-time PCR and Western blot. Results: Only TFF3 but not TFF1 and -2 was expressed in SM from healthy donors as well as cases of OA or RA on protein and mRNA level. In contrast, all three TFFs were detected in all samples of SF on the protein level. No significant changes were observed for TFF1 at all. TFF2 was significantly upregulated in RA samples in comparison to OA samples. TFF3 protein was significantly downregulated in OA samples in comparison to healthy samples and cases of RA significantly upregulated compared to OA. In contrast, in SM TFF3 protein was not significantly regulated. Conclusion: The data demonstrate the production of TFF3 in SM. Unexpectedly, SF contains all three known TFF peptides. As neither articular cartilage nor SM produce TFF1 and TFF2, we speculate that these originate with high probability from blood serum. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Figure 1

9 pages, 1119 KiB  
Article
The TFF Peptides xP1 and xP4 Appear in Distinctive Forms in the Xenopus laevis Gastric Mucosa: Indications for Different Protective Functions
by René Stürmer, Jana Reising and Werner Hoffmann
Int. J. Mol. Sci. 2019, 20(23), 6052; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20236052 - 30 Nov 2019
Cited by 14 | Viewed by 2024
Abstract
The gastric secretory trefoil factor family (TFF) peptides xP1 and xP4 are the Xenopus laevis orthologs of mammalian TFF1 and TFF2, respectively. The aim of this study was to analyze the molecular forms of xP1 and xP4 in the X. laevis gastric mucosa [...] Read more.
The gastric secretory trefoil factor family (TFF) peptides xP1 and xP4 are the Xenopus laevis orthologs of mammalian TFF1 and TFF2, respectively. The aim of this study was to analyze the molecular forms of xP1 and xP4 in the X. laevis gastric mucosa by FPLC. xP1 mainly occurred in a monomeric low-molecular-mass form and only a minor subset is associated with the mucus fraction. The occurrence of monomeric xP1 is unexpected because of its odd number of cysteine residues. Probably a conserved acidic residue flanking Cys55 allows monomeric secretion. Furthermore, Cys55 is probably post-translationally modified. For the first time, we hypothesize that the free thiol of monomeric xP1-and probably also its mammalian ortholog TFF1-could have a protective scavenger function, e.g., for reactive oxygen/nitrogen species. In contrast, xP4 mainly occurs in a high-molecular-mass form and is non-covalently bound to a mucin similarly as TFF2. In vitro binding studies with radioactively labeled porcine TFF2 even showed binding to X. laevis gastric mucin. Thus, xP4 is expected to bind as a lectin to an evolutionary conserved sugar epitope of the X. laevis ortholog of mucin MUC6 creating a tight mucus barrier. Taken together, xP1 and xP4 appear to have different gastric protective functions. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Graphical abstract

13 pages, 2254 KiB  
Article
Different Forms of TFF2, A Lectin of the Human Gastric Mucus Barrier: In Vitro Binding Studies
by Franziska Heuer, René Stürmer, Jörn Heuer, Thomas Kalinski, Antje Lemke, Frank Meyer and Werner Hoffmann
Int. J. Mol. Sci. 2019, 20(23), 5871; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20235871 - 22 Nov 2019
Cited by 19 | Viewed by 3612
Abstract
Trefoil factor family 2 (TFF2) and the mucin MUC6 are co-secreted from human gastric and duodenal glands. TFF2 binds MUC6 as a lectin and is a constituent of the gastric mucus. Herein, we investigated human gastric extracts by FPLC and identified mainly high- [...] Read more.
Trefoil factor family 2 (TFF2) and the mucin MUC6 are co-secreted from human gastric and duodenal glands. TFF2 binds MUC6 as a lectin and is a constituent of the gastric mucus. Herein, we investigated human gastric extracts by FPLC and identified mainly high- but also low-molecular-mass forms of TFF2. From the high-molecular-mass forms, TFF2 can be completely released by boiling in SDS or by harsh denaturing extraction. The low-molecular-mass form representing monomeric TFF2 can be washed out in part from gastric mucosa specimens with buffer. Overlay assays with radioactively labeled TFF2 revealed binding to the mucin MUC6 and not MUC5AC. This binding is modulated by Ca2+ and can be blocked by the lectin GSA-II and the monoclonal antibody HIK1083. TFF2 binding was also inhibited by Me-β-Gal, but not the α anomer. Thus, both the α1,4GlcNAc as well as the juxtaperipheral β-galactoside residues of the characteristic GlcNAcα1→4Galβ1→R moiety of human MUC6 are essential for TFF2 binding. Furthermore, there are major differences in the TFF2 binding characteristics when human is compared with the porcine system. Taken together, TFF2 appears to fulfill an important role in stabilizing the inner insoluble gastric mucus barrier layer, particularly by its binding to the mucin MUC6. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Graphical abstract

15 pages, 2279 KiB  
Article
Differential Expression of TFF1 and TFF3 in Patients Suffering from Chronic Rhinosinusitis with Nasal Polyposis
by Martina Mihalj, Maro Bujak, Josip Butković, Željko Zubčić, Maja Tolušić Levak, Josip Čes, Vlatko Kopić, Mirela Baus Lončar and Hrvoje Mihalj
Int. J. Mol. Sci. 2019, 20(21), 5461; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20215461 - 01 Nov 2019
Cited by 7 | Viewed by 2687
Abstract
Trefoil family factor (TFF) proteins contribute to antimicrobial defense and the maintenance of sinonasal epithelial barrier integrity. Dysregulation of TFF expression may be involved in the development of chronic inflammation and tissue remodeling characteristically found in chronic rhinosinusitis with nasal polyposis (CRSwNP). Expressions [...] Read more.
Trefoil family factor (TFF) proteins contribute to antimicrobial defense and the maintenance of sinonasal epithelial barrier integrity. Dysregulation of TFF expression may be involved in the development of chronic inflammation and tissue remodeling characteristically found in chronic rhinosinusitis with nasal polyposis (CRSwNP). Expressions of TFF1 and TFF3 were determined in specimens of middle nasal turbinate (MNT-0), bulla ethmoidalis (BE), and nasal polyps (NP) from CRSwNP patients (n = 29) and inferior nasal turbinate from a group of control patients (underwent nasal septoplasty, n = 25). An additional MNT sample was collected 6 months after functional endoscopic sinus surgery (FESS, MNT-6). TFF1 mRNA levels were significantly reduced in all specimens by approximately three- to five-fold, while TFF3 was increased in MNT-0, as compared with controls. Six months after surgery their levels were reversed to control values. CRSwNP patients with S. epidermidis isolated from sinus swabs showed upregulation of TFF3 in MNT and NP as compared with patients with sterile swabs. Target gene regulation was not affected by the presence of type 2 inflammation in patients with confirmed allergy. Results of this study imply participation of TFFs genes in the development of CRSwNP. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Figure 1

25 pages, 7309 KiB  
Article
Impact of High Salt Diet on Cerebral Vascular Function and Stroke in Tff3−/−/C57BL/6N Knockout and WT (C57BL/6N) Control Mice
by Nataša Kozina, Zrinka Mihaljević, Mirela Baus Lončar, Martina Mihalj, Mihael Mišir, Marina Dobrivojević Radmilović, Helena Justić, Srećko Gajović, Kate Šešelja, Iva Bazina, Anita Horvatić, Anita Matić, Nikola Bijelić, Edi Rođak, Ivana Jukić and Ines Drenjančević
Int. J. Mol. Sci. 2019, 20(20), 5188; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20205188 - 19 Oct 2019
Cited by 7 | Viewed by 4226
Abstract
High salt (HS) dietary intake leads to impaired vascular endothelium-dependent responses to various physiological stimuli, some of which are mediated by arachidonic acid (AA) metabolites. Transgenic Tff3−/− gene knockout mice (Tff3−/−/C57BL/6N) have changes in lipid metabolism which may [...] Read more.
High salt (HS) dietary intake leads to impaired vascular endothelium-dependent responses to various physiological stimuli, some of which are mediated by arachidonic acid (AA) metabolites. Transgenic Tff3−/− gene knockout mice (Tff3−/−/C57BL/6N) have changes in lipid metabolism which may affect vascular function and outcomes of stroke. We aimed to study the effects of one week of HS diet (4% NaCl) on vascular function and stroke induced by transient occlusion of middle cerebral artery in Tff3−/− and wild type (WT/C57BL/6N) mice. Flow-induced dilation (FID) of carotid artery was reduced in WT-HS mice, but not affected in Tff3−/−-HS mice. Nitric oxide (NO) mediated FID. NO production was decreased with HS diet. On the contrary, acetylcholine-induced dilation was significantly decreased in Tff3−/− mice on both diets and WT-HS mice. HS intake and Tff3 gene depletion affected the structural components of the vessels. Proteomic analysis revealed a significant effect of Tff3 gene deficiency on HS diet-induced changes in neuronal structural proteins and acute innate immune response proteins’ expression and Tff3 depletion, but HS diet did not increase the stroke volume, which is related to proteome modification and upregulation of genes involved mainly in cellular antioxidative defense. In conclusion, Tff3 depletion seems to partially impair vascular function and worsen the outcomes of stroke, which is moderately affected by HS diet. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Figure 1

10 pages, 1353 KiB  
Article
Different Forms of TFF3 in the Human Saliva: Heterodimerization with IgG Fc Binding Protein (FCGBP)
by Till Houben, Sönke Harder, Harmut Schlüter, Hubert Kalbacher and Werner Hoffmann
Int. J. Mol. Sci. 2019, 20(20), 5000; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20205000 - 10 Oct 2019
Cited by 27 | Viewed by 3064
Abstract
The peptide TFF3 is a member of a family of secretory lectins, and is typically synthesized by mucous epithelia together with mucins. It is mainly released from intestinal goblet cells as a high-molecular mass heterodimer with IgG Fc binding protein (FCGBP). Herein, we [...] Read more.
The peptide TFF3 is a member of a family of secretory lectins, and is typically synthesized by mucous epithelia together with mucins. It is mainly released from intestinal goblet cells as a high-molecular mass heterodimer with IgG Fc binding protein (FCGBP). Herein, we investigated human saliva by fast protein liquid chromatography (FPLC) and proteomics and identified high- and low-molecular-mass forms of TFF3. Whereas the high-molecular-mass forms represent a heterodimer with FCGBP, the low-molecular-mass forms represent homodimeric TFF3 forms. Proteomic analysis also revealed a C-terminally truncated form of TFF3. We hypothesize that salivary TFF3-FCGBP might play a role in the innate immune defense of the oral cavity and that TFF3 might also bind to microbial glycans. The known interaction of TFF3 with the agglutinin DMBT-1, a typical constituent of human saliva, further supports this protective role. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Graphical abstract

19 pages, 3926 KiB  
Article
Effect of Tff3 Deficiency and ER Stress in the Liver
by Kate Šešelja, Iva Bazina, Jessica Welss, Martin Schicht, Friedrich Paulsen, Nikola Bijelić, Edi Rođak, Anita Horvatić, Andrea Gelemanović, Martina Mihalj and Mirela Baus Lončar
Int. J. Mol. Sci. 2019, 20(18), 4389; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20184389 - 06 Sep 2019
Cited by 8 | Viewed by 4798
Abstract
Endoplasmic reticulum (ER) stress, a cellular condition caused by the accumulation of unfolded proteins inside the ER, has been recognized as a major pathological mechanism in a variety of conditions, including cancer, metabolic and neurodegenerative diseases. Trefoil factor family (TFFs) peptides are present [...] Read more.
Endoplasmic reticulum (ER) stress, a cellular condition caused by the accumulation of unfolded proteins inside the ER, has been recognized as a major pathological mechanism in a variety of conditions, including cancer, metabolic and neurodegenerative diseases. Trefoil factor family (TFFs) peptides are present in different epithelial organs, blood supply, neural tissues, as well as in the liver, and their deficiency has been linked to the ER function. Complete ablation of Tff3 expression is observed in steatosis, and as the most prominent change in the early phase of diabetes in multigenic mouse models of diabesity. To elucidate the role of Tff3 deficiency on different pathologically relevant pathways, we have developed a new congenic mouse model Tff3−/−/C57BL6/N from a mixed background strain (C57BL6/N /SV129) by using a speed congenics approach. Acute ER stress was evoked by tunicamycin treatment, and mice were sacrificed after 24 h. Afterwards the effect of Tff3 deficiency was evaluated with regard to the expression of relevant oxidative and ER stress genes, relevant proinflammatory cytokines/chemokines, and the global protein content. The most dramatic change was noticed at the level of inflammation-related genes, while markers for unfolded protein response were not significantly affected. Ultrastructural analysis confirmed that the size of lipid vacuoles was affected as well. Since the liver acts as an important metabolic and immunological organ, the influence of Tff3 deficiency and physiological function possibly reflects on the whole organism. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Figure 1

13 pages, 2141 KiB  
Article
Trefoil Factor 3 (TFF3) Is Involved in Cell Migration for Skeletal Repair
by Katharina Krüger, Sebastian Schmid, Friedrich Paulsen, Anita Ignatius, Patricia Klinger, Thilo Hotfiel, Bernd Swoboda and Kolja Gelse
Int. J. Mol. Sci. 2019, 20(17), 4277; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20174277 - 01 Sep 2019
Cited by 9 | Viewed by 2727
Abstract
The aim of the study was to explore the possible role of Trefoil Factor Family peptide 3 (TFF3) for skeletal repair. The expression of TFF3 was analyzed in human joint tissues as well as in a murine bone fracture model. Serum levels of [...] Read more.
The aim of the study was to explore the possible role of Trefoil Factor Family peptide 3 (TFF3) for skeletal repair. The expression of TFF3 was analyzed in human joint tissues as well as in a murine bone fracture model. Serum levels of TFF3 following a defined skeletal trauma in humans were determined by ELISA. The mRNA expression of TFF3 was analyzed under normoxia and hypoxia. Expression analysis after stimulation of human mesenchymal progenitor cells (MPCs) with TFF3 was performed by RT2 Profiler PCR Array. The effect of recombinant human (rh)TFF3 on MPCs was analysed by different migration and chemotaxis assays. The effect on cell motility was also visualized by fluorescence staining of F-Actin. TFF3 was absent in human articular cartilage, but strongly expressed in the subchondral bone and periosteum of adult joints. Strong TFF3 immunoreactivity was also detected in murine fracture callus. Serum levels of TFF3 were significantly increased after skeletal trauma in humans. Expression analysis demonstrated that rhTFF3 significantly decreased mRNA of ROCK1. Wound healing assays showed increased cell migration of MPCs by rhTFF3. The F-Actin cytoskeleton was markedly influenced by rhTFF3. Cell proliferation was not increased by rhTFF3. The data demonstrate elevated expression of TFF3 after skeletal trauma. The stimulatory effects on cell motility and migration of MPCs suggest a role of TFF3 in skeletal repair. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Figure 1

15 pages, 15408 KiB  
Article
p53, miR-34a and EMP1—Newly Identified Targets of TFF3 Signaling in Y79 Retinoblastoma Cells
by Maike Busch, Stefan Klein, Jan Große-Kreul, Oliver Scheiner, Klaus Metz, Harald Stephan and Nicole Dünker
Int. J. Mol. Sci. 2019, 20(17), 4129; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20174129 - 24 Aug 2019
Cited by 9 | Viewed by 3886
Abstract
Trefoil factor family peptide 3 (TFF3) is supposed to have tumor suppressive functions in retinoblastoma (RB), but the functional pathway is not completely understood. In the study presented, we investigated the downstream pathway of TFF3 signaling in Y79 RB cells. Results from pG13-luciferase [...] Read more.
Trefoil factor family peptide 3 (TFF3) is supposed to have tumor suppressive functions in retinoblastoma (RB), but the functional pathway is not completely understood. In the study presented, we investigated the downstream pathway of TFF3 signaling in Y79 RB cells. Results from pG13-luciferase reporter assays and western blot analyses indicate induced p53 activity with an upregulation of miR-34a after TFF3 overexpression. Expression levels of the predicted miR-34a target epithelial membrane protein 1 (EMP1) are reduced after TFF3 overexpression. As revealed by WST-1 assay, BrdU, and DAPI cell counts viability and proliferation of Y79 cells significantly decrease following EMP1 knockdown, while apoptosis levels significantly increase. Opposite effects on Y79 cells’ growth could be shown after EMP1 overexpression. Caspase assays showed that EMP1 induced apoptosis after overexpression is at least partially caspase-3/7 dependent. Colony formation and soft agarose assays, testing for anchorage independent growth, revealed that EMP1 overexpressing Y79 cells have a significantly higher ability to form colonies. In in ovo chicken chorioallantoic membrane (CAM) assays inoculated EMP1 overexpressing Y79 cells form significantly larger CAM tumors. Moreover, miR-34a overexpression increases sensitivity of Y79 cells towards RB chemotherapeutics, however, without involvement of EMP1. In summary, the TFF3 signaling pathway in Y79 RB cells involves the activation of p53 with downstream induction of miR-34a and subsequent inhibition of EMP1. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Figure 1

20 pages, 5372 KiB  
Article
Probiotics Upregulate Trefoil Factors and Downregulate Pepsinogen in the Mouse Stomach
by Ghalia Khoder, Farah Al-Yassir, Asma Al Menhali, Prashanth Saseedharan, Subi Sugathan, Catherine Tomasetto and Sherif M. Karam
Int. J. Mol. Sci. 2019, 20(16), 3901; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20163901 - 10 Aug 2019
Cited by 13 | Viewed by 3571
Abstract
Probiotics are used in the management of some gastrointestinal diseases. However, little is known about their effects on normal gastric epithelial biology. The aim of this study was to explore how the probiotic mixture VSL#3 affects gastric cell lineages in mice with a [...] Read more.
Probiotics are used in the management of some gastrointestinal diseases. However, little is known about their effects on normal gastric epithelial biology. The aim of this study was to explore how the probiotic mixture VSL#3 affects gastric cell lineages in mice with a special focus on protective and aggressive factors. Weight-matching littermate male mice (n = 14) were divided into treated and control pairs. The treated mice received VSL#3 (5 mg/day/mouse) by gastric gavage for 10 days. Control mice received only the vehicle. Food consumption and bodyweight were monitored. All mice were injected intraperitoneally with bromodeoxyuridine (120 mg/Kg bodyweight) two hours before sacrificed to label S-phase cells. Stomach tissues were processed for lectin- and immunohistochemical examination. ImageJ software was used to quantify immunolabeled gastric epithelial cells. Real-time quantitative polymerase chain reaction was used to provide relative changes in expression of gastric cell lineages specific genes. Results revealed that treated mice acquired (i) increased production of mucus, trefoil factor (TFF) 1 and TFF2, (ii) decreased production of pepsinogen, and (iii) increased ghrelin-secreting cells. No significant changes were observed in bodyweight, food consumption, cell proliferation, or parietal cells. Therefore, VSL#3 administration amplifies specific cell types specialized in the protection of the gastric epithelium. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Figure 1

Review

Jump to: Research

14 pages, 3697 KiB  
Review
The Interaction of Helicobacter pylori with TFF1 and Its Role in Mediating the Tropism of the Bacteria Within the Stomach
by Marguerite Clyne and Felicity E. B. May
Int. J. Mol. Sci. 2019, 20(18), 4400; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20184400 - 07 Sep 2019
Cited by 20 | Viewed by 4230
Abstract
Helicobacter pylori colonises the human stomach and has tropism for the gastric mucin, MUC5AC. The majority of organisms live in the adherent mucus layer within their preferred location, close to the epithelial surface where the pH is near neutral. Trefoil factor 1 (TFF1) [...] Read more.
Helicobacter pylori colonises the human stomach and has tropism for the gastric mucin, MUC5AC. The majority of organisms live in the adherent mucus layer within their preferred location, close to the epithelial surface where the pH is near neutral. Trefoil factor 1 (TFF1) is a small trefoil protein co-expressed with the gastric mucin MUC5AC in surface foveolar cells and co-secreted with MUC5AC into gastric mucus. Helicobacter pylori binds with greater avidity to TFF1 dimer, which is present in gastric mucus, than to TFF1 monomer. Binding of H. pylori to TFF1 is mediated by the core oligosaccharide subunit of H. pylori lipopolysaccharide at pH 5.0–6.0. Treatment of H. pylori lipopolysaccharide with mannosidase or glucosidase inhibits its interaction with TFF1. Both TFF1 and H. pylori have a propensity for binding to mucins with terminal non-reducing α- or β-linked N-acetyl-d-glucosamine or α-(2,3) linked sialic acid or Gal-3-SO42−. These findings are strong evidence that TFF1 has carbohydrate-binding properties that may involve a conserved patch of aromatic hydrophobic residues on the surface of its trefoil domain. The pH-dependent lectin properties of TFF1 may serve to locate H. pylori deep in the gastric mucus layer close to the epithelium rather than at the epithelial surface. This restricted localisation could limit the interaction of H. pylori with epithelial cells and the subsequent host signalling events that promote inflammation. Full article
(This article belongs to the Special Issue TFF Peptides: Lectins in Mucosal Protection and More)
Show Figures

Figure 1

Back to TopTop