Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 54
1.
Sci Rep ; 14(1): 10340, 2024 05 06.
Article En | MEDLINE | ID: mdl-38710764

This study aims to evaluate the role of trefoil factor 3 (TFF3) peptides in type 2 diabetes mellitus (T2DM) from an inflammatory perspective. The focus was on exploring how TFF3 affects the function of T cells. TFF3 overexpression model was constructed using lentivirus in Jurkat cell lines. We evaluated the impact of TFF3 on the proliferation, apoptosis, and IL-17A levels of Jurkat cells cultured in high glucose. The T2DM model was induced in TFF3 knockout (KO) mice through streptozotocin combined with high-fat diet. The measurements included glucose tolerance, insulin tolerance, inflammation markers, Th17 cell proportion, and pancreatic pathological changes. The T2DM modeling led to splenomegaly in mice, and increased expression of TFF3 in their spleens. Overexpression of TFF3 increased the proportion of IL-17+ T cells and the levels of Th17-related cytokines in Jurkat cells. There was no difference in body weight and blood glucose levels between wild-type and TFF3 KO mice. However, T2DM mice lacking the TFF3 gene showed improved glucose utilization, ameliorated pancreatic pathology, decreased inflammation levels, and reduced Th17 cell ratio. TFF3 may be involved in the chronic inflammatory immune response in T2DM. Its mechanism may be related to the regulation of the RORγt/IL-17 signaling pathway and its impact on T cell proliferation and apoptosis.


Diabetes Mellitus, Type 2 , Mice, Knockout , Th17 Cells , Trefoil Factor-3 , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Humans , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/immunology , Mice , Trefoil Factor-3/metabolism , Trefoil Factor-3/genetics , Jurkat Cells , Interleukin-17/metabolism , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Male , Cell Proliferation , Apoptosis , Diet, High-Fat/adverse effects
2.
Int J Mol Sci ; 25(4)2024 Feb 14.
Article En | MEDLINE | ID: mdl-38396964

TFF3 is a typical secretory poplypeptide of mucous epithelia belonging to the trefoil factor family (TFF) of lectins. In the intestine, respiratory tract, and saliva, TFF3 mainly exists as a high-molecular-mass complex with IgG Fc binding protein (FCGBP), which is indicative of a role in mucosal innate immunity. For the first time, we identified different forms of TFF3 in the endocervix, i.e., monomeric and homodimeric TFF3, as well as a high-molecular-mass TFF3-FCGBP complex; the latter also exists in a hardly soluble form. Immunohistochemistry co-localized TFF3 and FCGBP. Expression analyses of endocervical and post-menopausal vaginal specimens revealed a lack of mucin and TFF3 transcripts in the vaginal specimens. In contrast, genes encoding other typical components of the innate immune defense were expressed in both the endocervix and vagina. Of note, FCGBP is possibly fucosylated. Endocervical specimens from transgender individuals after hormonal therapy showed diminished expression, particularly of FCGBP. Furthermore, mucus swabs from the endocervix and vagina were analyzed concerning TFF3, FCGBP, and lysozyme. It was the aim of this study to illuminate several aspects of the cervico-vaginal innate immune barrier, which is clinically relevant as bacterial and viral infections are also linked to infertility, pre-term birth and cervical cancer.


Cervix Uteri , Mucins , Vagina , Female , Humans , Carrier Proteins , Cell Adhesion Molecules/metabolism , Cervix Uteri/immunology , Immunity, Innate , Immunoglobulin G/metabolism , Mucins/metabolism , Trefoil Factor-2/metabolism , Trefoil Factor-3/genetics , Trefoil Factor-3/metabolism , Vagina/immunology
3.
Int J Mol Sci ; 24(22)2023 Nov 15.
Article En | MEDLINE | ID: mdl-38003531

Trefoil factor family protein 3 (Tff3) protects the gastrointestinal mucosa and has a complex mode of action in different tissues. Here, we aimed to determine the effect of Tff3 deficiency on intestinal tissues in a long-term high-fat-diet (HFD)-fed model. A novel congenic strain without additional metabolically relevant mutations (Tff3-/-/C57Bl6NCrl strain, male and female) was used. Wild type (Wt) and Tff3-deficient mice of both sexes were fed a HFD for 36 weeks. Long-term feeding of a HFD induces different effects on the intestinal structure of Tff3-deficient male and female mice. For the first time, we found sex-specific differences in duodenal morphology. HFD feeding reduced microvilli height in Tff3-deficient females compared to that in Wt females, suggesting a possible effect on microvillar actin filament dynamics. These changes could not be attributed to genes involved in ER and oxidative stress, apoptosis, or inflammation. Tff3-deficient males exhibited a reduced cecal crypt depth compared to that of Wt males, but this was not the case in females. Microbiome-related short-chain fatty acid content was not affected by Tff3 deficiency in HFD-fed male or female mice. Sex-related differences due to Tff3 deficiency imply the need to consider both sexes in future studies on the role of Tff in intestinal function.


Diet, High-Fat , Proteins , Mice , Male , Animals , Female , Diet, High-Fat/adverse effects , Mice, Inbred Strains , Duodenum , Mice, Inbred C57BL , Trefoil Factor-3/genetics
4.
Nutrients ; 15(11)2023 May 24.
Article En | MEDLINE | ID: mdl-37299399

Trefoil factor 3 (TFF3) plays a key role in the maintenance and repair of intestinal mucosa. TFF3 expression is upregulated by the microbiota through TLR2. At the posttranscriptional level, TFF3 is downregulated by miR-7-5p. Reduced TFF3 levels have been detected in the damaged tissue of IBD patients. Here, we investigate the regulation of TFF3 expression by microbiota extracellular vesicles (EVs) in LS174T goblet cells using RT-qPCR and inhibitors of the TLR2 or PI3K pathways. To evaluate the subsequent impact on epithelial barrier function, conditioned media from control and vesicle-stimulated LS174T cells were used to treat Caco-2 monolayers. The barrier-strengthening effects were evaluated by analysing the expression and subcellular distribution of tight junction proteins, and the repairing effects were assessed using wound-healing assays. The results showed a differential regulation of TFF3 in LS174T via EVs from the probiotic EcN and the commensal ECOR12. EcN EVs activated the TFF3 production through TLR2 and downregulated miR7-5-p through PI3K. Consistently, high levels of secreted TFF3 reinforced the tight junctions and stimulated wound healing in the Caco-2 cells. ECOR12 EVs did not cause these effects. TFF3 is a potential therapeutic target in IBD. This study contributes to understanding the molecular players (microbiota EVs) connecting gut microbes to health and may help in designing better nutritional interventions based on microbiota bioactive compounds.


Extracellular Vesicles , Inflammatory Bowel Diseases , Humans , Goblet Cells/metabolism , Caco-2 Cells , Trefoil Factor-3/genetics , Trefoil Factor-3/metabolism , Trefoil Factor-3/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Toll-Like Receptor 2/metabolism , Intestinal Mucosa/metabolism , Inflammatory Bowel Diseases/metabolism , Extracellular Vesicles/metabolism
5.
Arch Biochem Biophys ; 729: 109379, 2022 10 30.
Article En | MEDLINE | ID: mdl-36002083

Obesity is a risk factor for gastrointestinal malignancies and tumors. However, which factors either protect or predispose the gastrointestinal organs to high-fat diet (HFD)-induced neoplasia remains unclear. Here, we demonstrate that HFD impacts the stomach to a greater extent as compared to the colorectum, resulting in leptin receptor (LepR) signaling-mediated neoplasia in the tissues. HFD activated leptin signaling, which in turn, accelerates the pathogenesis in the gastric mucosa more than that in the colorectum along with ectopic TFF3 expression. Moreover, in the stomach, higher levels of phosphorylated epidermal growth factor receptor (EGFR) in addition to the activation of STAT3 and Akt were observed as compared to the colorectum. The mice with LepR deletion in the gastrointestinal epithelium exhibited a suppressed induction of leptin, TFF3, and phosphorylated EGFR in the stomach, whereas the levels in the colorectum were insignificant. In co-transfected COS-7 cells with LepR and EGFR plasmid DNA, leptin transactivated EGFR to accelerate TFF3 induction along with activation of STAT3, ERK1/2, Akt, and PI3K p85/p55. Furthermore, TFF3 could bind to EGFR but did not transactivate LepR. Leptin-induced TFF3 induction was markedly suppressed by inhibitors of PI3K (LY294002) and EGFR (Erlotinib). Together, these results suggest a novel role of LepR-mediated signaling in transactivating EGFR that leads to TFF3 expression via the PI3K-Akt pathway. Therefore, this study sheds light on the identification of potentially new therapeutic targets for the treatment of pre-cancerous symptoms in stomach and colorectum.


Leptin , Receptors, Leptin , Animals , DNA , Dietary Fats/adverse effects , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Leptin/metabolism , Mice , Mice, Obese , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Stomach/pathology , Transcriptional Activation , Trefoil Factor-3/genetics , Trefoil Factor-3/metabolism
6.
Acta Neurobiol Exp (Wars) ; 82(2): 237-243, 2022.
Article En | MEDLINE | ID: mdl-35833823

Pituitary adenoma is a typical adult primary brain tumor and 35% of pituitary adenomas are invasive. The enhancement of angiogenesis is essential for the spread and invasiveness of invasive pituitary adenoma. Thus, it is urgent to uncover the mechanism and relevant biomolecular targets for the therapy and prognosis of pituitary adenomas. The HP75 cells were transfected with si­NC, si­TFF3, pcDNA, and pcDNA­TFF3 to investigate the effects of TFF3 on the proliferation, migration and invasion of pituitary tumor cell. The protein level of TFF3 and HIF­1α/VEGFA was determined by western blot. The transwell migration assay and wound healing assay were used to investigate the influence of TFF3 on the cell migration and invasion of HP75 cells. The tumor angiogenesis was determined by tube formation assay. The proliferation of HP75 cells was assessed by using MTT assay and colony­forming unit assay. The cell proliferation rate was separately enhanced and reduced remarkably in TFF3 overexpression group and si­TFF3 group. TFF3 could modulate the proliferation, migration and invasion ability of HP75 cells. Furthermore, TFF3 may play a oncogenic role in HP75 cells. Overexpression of TFF3 enhanced the number of branching points and network formation in HP75 cells, suggesting the TFF3 had positive effects on cell angiogenesis. These results also disclosed a novel relationship between TFF3 expression and the activation of the HIF­1α/VEGFA signaling pathway. In summary, this study uncovered new insight into the mechanisms of TFF3's anti­tumor activities in pituitary adenoma cells by investigating its effects on HIF­1α/VEGFA signaling pathway regulation.


Adenoma , Neovascularization, Pathologic , Pituitary Neoplasms , Trefoil Factor-3 , Vascular Endothelial Growth Factor A , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Adult , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Humans , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Signal Transduction , Trefoil Factor-3/genetics , Trefoil Factor-3/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
7.
J Clin Pharm Ther ; 47(5): 609-618, 2022 May.
Article En | MEDLINE | ID: mdl-35229335

WHAT IS KNOWN AND OBJECTIVE: Compared with other molecular subtypes, hormone receptor-positive breast cancer often shows worse neoadjuvant chemotherapy efficacy. This study aims to explore the relationship between the oestrogen receptor (ER)-related genes carbonic anhydrase 12 (CA12) and trefoil factor 3 (TFF3) and their predictive value of neoadjuvant chemotherapy for breast cancer. METHODS: We investigated the relationships between CA12, TFF3 and ER status and their predictive value of anthracycline-taxane neoadjuvant chemotherapy in 115 female breast cancer patients via real-time polymerase chain reaction (RT-PCR) and 4 GEO datasets: GSE41998, GSE25065, GSE20194 and GSE20271. Then, the effects of CA12 and TFF3 on the chemotherapy drugs doxorubicin and docetaxel were verified in vitro in the breast cancer cell lines MCF-7 and BT474. RESULTS AND DISCUSSION: The GEO datasets and RT-PCR results showed that the relative expression of both CA12 and TFF3 was higher in oestrogen receptor-positive samples compared with the other samples (p < 0.05). CA12 was significantly correlated with TFF3 (p < 0.05). In MCF-7 cells, inhibition of TFF3 induced downregulation of CA12 and ESR1 (p < 0.05) at both the mRNA and the protein levels, while inhibition of CA12 also downregulated TFF3 and ESR1 (p < 0.05). In BT474 cells, inhibition of TFF3 downregulated CA12 and ESR1 (p < 0.05) at both the mRNA and the protein levels, while inhibition of CA12 led to slight upregulation of TFF3 and ESR1 (p > 0.05). Moreover, GEO datasets and RT-PCR results showed that CA12 and TFF3 were more highly expressed in nonpathological complete response (non-pCR) samples than in pCR samples (p < 0.05). Cell viability assays of MCF-7 and BT474 cells showed that inhibiting CA12 and TFF3 could enhance sensitivity to doxorubicin and docetaxel (p < 0.05). WHAT IS NEW AND CONCLUSION: CA12 and TFF3 were correlated with each other, and their high expression might explain the worse efficacy of neoadjuvant chemotherapy in oestrogen receptor-positive breast cancer patients.


Breast Neoplasms , Neoadjuvant Therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carbonic Anhydrases , Docetaxel/pharmacology , Doxorubicin/therapeutic use , Female , Humans , RNA, Messenger , Receptors, Estrogen/metabolism , Receptors, Estrogen/therapeutic use , Trefoil Factor-3/genetics
8.
Cell Death Dis ; 13(1): 62, 2022 01 17.
Article En | MEDLINE | ID: mdl-35039476

Trefoil factor 3 (TFF3) is the last small-molecule peptide found in the trefoil factor family, which is mainly secreted by intestinal goblet cells and exerts mucosal repair effect in the gastrointestinal tract. Emerging evidence indicated that the TFF3 expression profile and biological effects changed significantly in pathological states such as cancer, colitis, gastric ulcer, diabetes mellitus, non-alcoholic fatty liver disease, and nervous system disease. More importantly, mucosal protection would no longer be the only effect of TFF3, it gradually exhibits carcinogenic activity and potential regulatory effect of nervous and endocrine systems, but the inner mechanisms remain unclear. Understanding the molecular function of TFF3 in specific diseases might provide a new insight for the clinical development of novel therapeutic strategies. This review provides an up-to-date overview of the pathological effects of TFF3 in different disease and discusses the binding proteins, signaling pathways, and clinical application.


Trefoil Factor-3 , Disease , Humans , Signal Transduction , Trefoil Factor-3/genetics
9.
Future Oncol ; 18(3): 333-348, 2022 Jan.
Article En | MEDLINE | ID: mdl-34756116

Background: Papillary thyroid carcinoma (PTC) is one of the most common endocrine malignancies and has a favorable prognosis. However, optimal treatments and prognostic markers have not been clearly identified. Methods: Gene expression data from primary PTC were downloaded from the Gene Expression Omnibus database and subjected to two analyses of differentially expressed genes (DEGs), followed by intersecting individual and integrated DEGs analyses as well as gene set enrichment analysis. Analysis of data from Sequence Read Archive and The Cancer Genome Atlas, immunohistochemistry and qRT-PCR of TFF3 were performed to validate the results. Finally, the relationship between gene expression and disease-free survival as well as immune cell infiltration were investigated. Results: Six critical DEGs and several tumor-enriched signaling pathways were identified. Immunohistochemistry and qRT-PCR validated the low expression of TFF3 in PTC. TFF3 and FCGBP are coexpressed in PTC, and patients with lower gene expression had worse disease-free survival but higher immune cell infiltration. Conclusion: TFF3 was significantly underexpressed and may function with FCGBP synergistically in PTC.


Lay abstract Thyroid cancers are some of the most common endocrine malignancies. However, the optimal treatments and prognostic markers have not been clearly identified. We identified six critical differentially expressed genes and several tumor-enriched signaling pathways in papillary thyroid carcinoma, and found that TFF3 was the most underexpressed gene, as validated by experiment. In addition, TFF3 and FCGBP worked synergistically and may mark prognosis and tumor immune cell infiltration, which may benefit patients with papillary thyroid carcinoma by providing early indication and prompting further basic investigation.


Biomarkers, Tumor/genetics , Neoplasm Recurrence, Local/epidemiology , Thyroid Cancer, Papillary/mortality , Thyroid Neoplasms/mortality , Trefoil Factor-3/genetics , Adult , Biomarkers, Tumor/analysis , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/genetics , Datasets as Topic , Disease-Free Survival , Female , Follow-Up Studies , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Risk Assessment/methods , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/immunology , Thyroid Cancer, Papillary/surgery , Thyroid Gland/immunology , Thyroid Gland/pathology , Thyroid Gland/surgery , Thyroid Neoplasms/genetics , Thyroid Neoplasms/immunology , Thyroid Neoplasms/surgery , Thyroidectomy , Trefoil Factor-3/analysis , Tumor Microenvironment/immunology
10.
Signal Transduct Target Ther ; 6(1): 268, 2021 07 14.
Article En | MEDLINE | ID: mdl-34262017

Major gaps in understanding the molecular mechanisms of colorectal cancer (CRC) progression and intestinal mucosal repair have hampered therapeutic development for gastrointestinal disorders. Trefoil factor 3 (TFF3) has been reported to be involved in CRC progression and intestinal mucosal repair; however, how TFF3 drives tumors to become more aggressive or metastatic and how TFF3 promotes intestinal mucosal repair are still poorly understood. Here, we found that the upregulated TFF3 in CRC predicted a worse overall survival rate. TFF3 deficiency impaired mucosal restitution and adenocarcinogenesis. CD147, a membrane protein, was identified as a binding partner for TFF3. Via binding to CD147, TFF3 enhanced CD147-CD44s interaction, resulting in signal transducer and activator of transcription 3 (STAT3) activation and prostaglandin G/H synthase 2 (PTGS2) expression, which were indispensable for TFF3-induced migration, proliferation, and invasion. PTGS2-derived PGE2 bound to prostaglandin E2 receptor EP4 subtype (PTGER4) and contributed to TFF3-stimulated CRC progression. Solution NMR studies of the TFF3-CD147 interaction revealed the key residues critical for TFF3 binding and the induction of PTGS2 expression. The ability of TFF3 to enhance mucosal restitution was weakened by a PTGS2 inhibitor. Blockade of TFF3-CD147 signaling using competitive inhibitory antibodies or a PTGS2 inhibitor reduced CRC lung metastasis in mice. Our findings bring strong evidence that CD147 is a novel receptor for TFF3 and PTGS2 signaling is critical for TFF3-induced mucosal restitution and CRC progression, which widens and deepens the understanding of the molecular function of trefoil factors.


Basigin/genetics , Colorectal Neoplasms/drug therapy , Cyclooxygenase 2/genetics , Receptors, Prostaglandin E, EP4 Subtype/genetics , Trefoil Factor-3/genetics , Animals , Basigin/antagonists & inhibitors , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cyclooxygenase 2/drug effects , Disease Progression , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Protein Binding/drug effects , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
11.
Toxicol Appl Pharmacol ; 422: 115561, 2021 07 01.
Article En | MEDLINE | ID: mdl-33957193

Arsenic is a global health concern that causes toxicity through ingestion of contaminated water and food. In vitro studies suggest that arsenic reduces stem and progenitor cell differentiation. Thus, this study determined if arsenic disrupted intestinal stem cell (ISC) differentiation, thereby altering the number, location, and/or function of intestinal epithelial cells. Adult male C57BL/6 mice were exposed to 0 or 100 ppb sodium arsenite (AsIII) through drinking water for 5 weeks. Duodenal sections were collected to assess changes in morphology, proliferation, and cell types. qPCR analysis revealed a 40% reduction in Lgr5 transcripts, an ISC marker, in the arsenic-exposed mice, although there were no changes in the protein expression of Olfm4. Secretory cell-specific transcript markers of Paneth (Defa1), Goblet (Tff3), and secretory transit amplifying (Math1) cells were reduced by 51%, 44%, and 30% respectively, in the arsenic-exposed mice, indicating significant impacts on the Wnt-dependent differentiation pathway. Further, protein levels of phosphorylated ß-catenin were reduced in the arsenic-exposed mice, which increased the expression of Wnt-dependent transcripts CD44 and c-myc. PCA analysis, followed by MANOVA and regression analyses, revealed significant changes and correlations between Lgr5 and the transit amplifying (TA) cell markers Math1 and Hes1, which are in the secretory cell pathway. Similar comparisons between Math1 and Defa1 show that terminal differentiation into Paneth cells is also reduced in the arsenic-exposed mice. The data suggests that ISCs are not lost following arsenic exposure, but rather, specific Wnt-dependent progenitor cell formation and terminal differentiation in the small intestine is reduced.


Arsenites/toxicity , Cell Differentiation/drug effects , Duodenum/drug effects , Paneth Cells/drug effects , Receptors, G-Protein-Coupled/metabolism , Sodium Compounds/toxicity , Stem Cells/drug effects , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Down-Regulation , Duodenum/metabolism , Duodenum/pathology , Male , Mice, Inbred C57BL , Paneth Cells/metabolism , Paneth Cells/pathology , Receptors, G-Protein-Coupled/genetics , Stem Cells/metabolism , Stem Cells/pathology , Trefoil Factor-3/genetics , Trefoil Factor-3/metabolism , Wnt Signaling Pathway , alpha-Defensins/genetics , alpha-Defensins/metabolism
12.
Eur J Immunol ; 51(5): 1110-1125, 2021 05.
Article En | MEDLINE | ID: mdl-33547649

Intestinal trefoil factor 3 (TFF3) plays an important role in repairing the intestinal mucosa. However, the detailed mechanism regarding immune regulation by TFF3 is not well defined. Here, we reported that treatment of mouse BM cells and human peripheral blood mononuclear cells from healthy volunteers with TFF3 activated polymorphnuclear myeloid-derived suppressor cells (PMN-MDSCs) in vitro. We also found that prostaglandin E2 is a major TFF3-mediated MDSC target, and that NF-κB/COX2 signaling was involved in this process. Moreover, TFF3 treatment or transfer of TFF3-derived PMN-MDSCs (TFF3-MDSCs) to experimental necrotizing enterocolitis (NEC) mice caused PMN-MDSC accumulation in the lamina propria (LP), which was associated with decreased intestinal inflammation, permeability, bacterial loading, and prolonged survival. Interestingly, no NEC severity remission was observed in Rag1 KO mice that were given TFF3-MDSCs, but coinjection with CD4+ T cells significantly relieved NEC inflammation. Overall, TFF3 mediates the NF-κB/COX2 pathway to regulate PMN-MDSC activation and attenuates NEC in a T-cell-dependent manner, which suggests a novel mechanism in preventing NEC occurrence.


Cyclooxygenase 2/metabolism , Enterocolitis, Necrotizing/etiology , Enterocolitis, Necrotizing/metabolism , Myeloid-Derived Suppressor Cells/metabolism , NF-kappa B/metabolism , Neutrophils/metabolism , Signal Transduction , Trefoil Factor-3/genetics , Animals , Animals, Newborn , Dinoprostone/metabolism , Disease Models, Animal , Disease Susceptibility , Enterocolitis, Necrotizing/pathology , Gene Expression Regulation , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Myeloid-Derived Suppressor Cells/immunology , Neutrophils/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Trefoil Factor-3/metabolism
13.
Biomarkers ; 26(2): 95-102, 2021 Mar.
Article En | MEDLINE | ID: mdl-33401971

OBJECTIVE: To assess the diagnostic value of Trefoil factor 3 (TFF3) and the correlation between TFF3 expression and clinicopathological features in patients with gastric cancer (GC). METHODS: PubMed, The Cochrane, EMbase, and Web of Science were retrieved comprehensively to collect relevant literature. The search ended on 31 May 2020. All data were analyzed using PubMed, The Cochrane, EMbase, and Web of Science were retrieved to collect relevant articles. All data from the included studies were subjected to meta-analysis using Stata 12.0 software. RESULTS: Seventeen studies involved 4654 subjects were included. For the diagnostic value of TFF3 for GC, the sensitivity, specificity, and AUC were 0.71, 0.80, and 0.80, respectively. For the clinicopathological value of TFF3, tissue TFF3 expression showed a higher risk of lymph node metastasis (OR 2.20, 95% CI 1.75-2.78, p < 0.001) and muscularis propria invasion (≥T2) (1.51, 1.13-2.03, p = 0.006), as well as worse TNM stage (2.26, 1.63-3.12, p < 0.001) and histological type (1.72, 1.34-2.20, p < 0.001), while no apparent relationship was found for serous membrane invasion (T4), venous invasion, and peritoneal metastasis. CONCLUSION: TFF3 may be a promising biomarker for GC, and the TFF3 expression is likely to be involved in the invasion, metastasis, and differentiation of GC.


Biomarkers, Tumor/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Trefoil Factor-3/genetics , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/genetics , Aged , Area Under Curve , Biomarkers, Tumor/metabolism , Case-Control Studies , Female , Gene Expression , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Sensitivity and Specificity , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Trefoil Factor-3/metabolism , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/secondary
14.
Oncol Rep ; 45(1): 254-264, 2021 01.
Article En | MEDLINE | ID: mdl-33210724

In colorectal cancer (CRC), high expression of trefoil factor 3 (TFF3) is associated with tumor progression and reduced patient survival; however, bioinformatics analyses of public 'omics' databases show low TFF3 expression in CRCs as compared to normal tissues. Thus, we examined TFF3 expression in CRCs and matching normal tissues to evaluate its role in CRC progression. TFF3 gene expression was characterized using the bioinformatics portal UALCAN (http://ualcan.path.uab.edu). Tissue microarrays (TMAs) of archival CRC specimens (n=96) were immunostained with anti­human TFF3 antibodies. Immunohistochemical (IHC) staining intensity was semi­quantitatively scored. For this cohort, the median follow­up was 5.4 years. Associations between clinical and pathological variables were determined using Chi­square or Fisher's exact tests. Univariate disease­free survival was estimated by the Kaplan­Meier method. Omics data analyses by UALCAN showed downregulation of TFF3 expression in CRC relative to normal tissue at protein (χ2, P<0.0001) levels. There was a similar decreasing trend of TFF3 expression in the pathologic stages of the CRCs (RNA, χ2, P=0.88 and protein, χ2 P<0.0001). UALCAN data analysis showed that TFF3 exhibited 27% lower mRNA expression in tumors with mutant TP53 (P=0.007). Confirming the findings of omics analyses, IHC analysis of TMAs exhibited lower TFF3 expression in 95.6% (65 of 68) of the available normal­tumor matching pairs (χ2, P<0.0001). There was no statistically significant association of tumor TFF3 expression with patient sex, race/ethnicity, tumor location within the colorectum, Tumor, Node, Metastasis (TNM) stage, lymph node metastasis, or surgical margins. However, low TFF3 IHC staining in tumor tissue was associated with histological grade (P=0.026). Kaplan­Meier survival analysis showed no prognostic value of low TFF3 expression relative to those with high expression (log­rank, P=0.605). Our findings demonstrate low expression of TFF3 in CRCs. Association between low TFF3 and histopathological features suggests involvement of this molecule in progression of CRC.


Colorectal Neoplasms/chemistry , Trefoil Factor-3/analysis , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Computational Biology , Female , Humans , Male , Middle Aged , Mutation , Trefoil Factor-3/genetics , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/genetics
15.
J Cell Mol Med ; 24(15): 8589-8602, 2020 08.
Article En | MEDLINE | ID: mdl-32652877

Chemoresistance represents a major challenge in breast cancer (BC) treatment. This study aimed to probe the roles of LINC00160 in paclitaxel- and doxorubicin-resistant BC cells. Three pairs of BC and adjacent normal tissue were used for lncRNA microarray analysis. Paclitaxel-resistant MCF-7 (MCF-7/Tax) and doxorubicin-resistant BT474 (BT474/Dox) cells were generated by exposure of parental drug-sensitive MCF-7 or BT474 cells to gradient concentrations of drugs. Correlation between LINC00160 expression and clinical response to paclitaxel in BC patients was examined. Short interfering RNAs specifically targeting LINC00160 or TFF3 were designed to construct LINC00160- and TFF3-depleted BC cells to discuss their effects on biological episodes of MCF-7/Tax and BT474/Dox cells. Interactions among LINC00160, transcription factor C/EBPß and TFF3 were identified. MCF-7/Tax and BT474/Dox cells stable silencing of LINC00160 were transplanted into nude mice. Consequently, up-regulated LINC00160 led to poor clinical response to paclitaxel in BC patients. LINC00160 knockdown reduced drug resistance in MCF-7/Tax and BT474/Dox cells and reduced cell migration and invasion. LINC00160 recruited C/EBPß into the promoter region of TFF3 and increased TFF3 expression. LINC00160-depleted MCF-7/Tax and BT474/Dox cells showed decreased tumour growth rates in nude mice. Overall, we identified a novel mechanism of LINC00160-mediated chemoresistance via the C/EBPß/TFF3 axis, highlighting the potential of LINC00160 for treating BC with chemoresistance.


CCAAT-Enhancer-Binding Protein-beta/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Paclitaxel/pharmacology , RNA, Long Noncoding , Trefoil Factor-3/genetics , Adult , Animals , Biomarkers, Tumor , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Humans , MCF-7 Cells , Mice , Middle Aged , Prognosis , RNA Interference , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
16.
EBioMedicine ; 58: 102877, 2020 Aug.
Article En | MEDLINE | ID: mdl-32707444

BACKGROUND: Radiofrequency ablation (RFA) is an effective treatment for dysplastic Barrett's esophagus (BE), but recurrence can occur after initial response. Currently there is uncertainty about how to best define histological remission. A DNA methylation panel on esophageal samples was previously shown to have high diagnostic accuracy for BE. We aimed to investigate this biomarker panel in the assessment of response to RFA treatment. METHODS: We retrospectively analyzed esophageal and gastroesophageal junction (GEJ) biopsies from patients with BE before and after RFA treatment. We quantified the extent of intestinal metaplasia (IM) based on number of glands with goblet cells (IM-Score) and expression of the intestinal factor trefoil factor-3 (TFF3-Score). Promoter methylation of 3 genes (ZNF345, TFP12, ZNF569) was measured by methylight (Meth-Score) throughout the RFA treatment pathway. FINDINGS: We included 45 patients (11 non-dysplastic BE, 14 low-grade dysplasia, 20 high-grade dysplasia/intramucosal cancer). Meth-Scores were significantly higher in BE with and without dysplasia and GEJ with IM compared to GEJ without IM (P<·001). Meth-scores significantly correlated with the extent of IM at the GEJ measured both with IM-Scores (rho=66·0%, P<·001), and TFF3-Scores (rho=75·6%, P<·001). In patients with residual IM at the GEJ, RFA re-treatment brought about a 7·6-fold reduction in the methylation levels. The Meth-score had an area under the ROC curve of 95·1% (95%CI 91·1% - 99·1%) differentiating BE from normal GEJ. INTERPRETATION: A DNA methylation panel can discriminate between the extent of histological IM in esophageal and junctional biopsies and could be used to objectively quantify residual disease following RFA.


Barrett Esophagus/pathology , Barrett Esophagus/therapy , Biomarkers, Tumor/genetics , DNA Methylation , Radiofrequency Ablation/methods , Aged , Barrett Esophagus/genetics , Biopsy , Case-Control Studies , Disease Progression , Epigenesis, Genetic , Female , Humans , Male , Middle Aged , Repressor Proteins/genetics , Retrospective Studies , Treatment Outcome , Trefoil Factor-3/genetics
17.
Nat Commun ; 11(1): 2265, 2020 05 13.
Article En | MEDLINE | ID: mdl-32404934

The mucosal epithelium secretes a host of protective disulfide-rich peptides, including the trefoil factors (TFFs). The TFFs increase the viscoelasticity of the mucosa and promote cell migration, though the molecular mechanisms underlying these functions have remained poorly defined. Here, we demonstrate that all TFFs are divalent lectins that recognise the GlcNAc-α-1,4-Gal disaccharide, which terminates some mucin-like O-glycans. Degradation of this disaccharide by a glycoside hydrolase abrogates TFF binding to mucins. Structural, mutagenic and biophysical data provide insights into how the TFFs recognise this disaccharide and rationalise their ability to modulate the physical properties of mucus across different pH ranges. These data reveal that TFF activity is dependent on the glycosylation state of mucosal glycoproteins and alludes to a lectin function for trefoil domains in other human proteins.


Lectins/metabolism , Mucus/metabolism , Trefoil Factor-1/metabolism , Trefoil Factor-3/metabolism , Crystallography, X-Ray , Disaccharides/metabolism , Glycoside Hydrolases/metabolism , Humans , Hydrogen Bonding , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Ligands , Mass Spectrometry , Mucins/metabolism , Phylogeny , Polysaccharides/metabolism , Trefoil Factor-1/chemistry , Trefoil Factor-1/genetics , Trefoil Factor-3/chemistry , Trefoil Factor-3/genetics
18.
Int J Mol Sci ; 20(23)2019 Dec 03.
Article En | MEDLINE | ID: mdl-31817054

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.


Arthritis, Rheumatoid/metabolism , Osteoarthritis/metabolism , Synovial Fluid/metabolism , Synovial Membrane/metabolism , Trefoil Factor-1/metabolism , Trefoil Factor-2/metabolism , Trefoil Factor-3/metabolism , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Peptides/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tissue Donors , Trefoil Factor-1/genetics , Trefoil Factor-2/genetics , Trefoil Factor-3/genetics
19.
Int J Mol Sci ; 20(21)2019 Nov 01.
Article En | MEDLINE | ID: mdl-31683988

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.


Nasal Polyps/genetics , Rhinitis/genetics , Sinusitis/genetics , Trefoil Factor-1/genetics , Trefoil Factor-3/genetics , Adult , Aged , Chronic Disease , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Middle Aged , Nasal Polyps/complications , Nasal Polyps/surgery , Reverse Transcriptase Polymerase Chain Reaction , Rhinitis/complications , Rhinitis/surgery , Sinusitis/complications , Sinusitis/surgery , Trefoil Factor-1/metabolism , Trefoil Factor-3/metabolism , Young Adult
20.
Nat Commun ; 10(1): 4408, 2019 09 27.
Article En | MEDLINE | ID: mdl-31562318

Intestinal epithelial cells (IEC) have important functions in nutrient absorption, barrier integrity, regeneration, pathogen-sensing, and mucus secretion. Goblet cells are a specialized cell type of IEC that secrete Trefoil factor 3 (TFF3) to regulate mucus viscosity and wound healing, but whether TFF3-responsiveness requires a receptor is unclear. Here, we show that leucine rich repeat receptor and nogo-interacting protein 2 (LINGO2) is essential for TFF3-mediated functions. LINGO2 immunoprecipitates with TFF3, co-localizes with TFF3 on the cell membrane of IEC, and allows TFF3 to block apoptosis. We further show that TFF3-LINGO2 interactions disrupt EGFR-LINGO2 complexes resulting in enhanced EGFR signaling. Excessive basal EGFR activation in Lingo2 deficient mice increases disease severity during colitis and augments immunity against helminth infection. Conversely, TFF3 deficiency reduces helminth immunity. Thus, TFF3-LINGO2 interactions de-repress inhibitory LINGO2-EGFR complexes, allowing TFF3 to drive wound healing and immunity.


Colitis/immunology , ErbB Receptors/immunology , Helminthiasis/immunology , Intestinal Mucosa/immunology , Membrane Proteins/metabolism , Nerve Tissue Proteins/immunology , Trefoil Factor-3/immunology , Animals , Cell Line, Tumor , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate , ErbB Receptors/genetics , ErbB Receptors/metabolism , Goblet Cells/immunology , Goblet Cells/metabolism , Goblet Cells/parasitology , HEK293 Cells , Helminthiasis/metabolism , Helminthiasis/parasitology , Helminths/immunology , Helminths/physiology , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Membrane Proteins/genetics , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organophosphonates , Trefoil Factor-3/genetics , Trefoil Factor-3/metabolism , U937 Cells
...