1
|
Lutz F, Han SY, Büyücek S, Möller K, Viehweger F, Schlichter R, Menz A, Luebke AM, Bawahab AA, Reiswich V, Kluth M, Hube-Magg C, Hinsch A, Weidemann S, Lennartz M, Dum D, Bernreuther C, Lebok P, Sauter G, Marx AH, Simon R, Krech T, Fraune C, Gorbokon N, Burandt E, Minner S, Steurer S, Clauditz TS, Jacobsen F. Expression of Trefoil Factor 1 (TFF1) in Cancer: A Tissue Microarray Study Involving 18,878 Tumors. Diagnostics (Basel) 2024; 14:2157. [PMID: 39410561 PMCID: PMC11475926 DOI: 10.3390/diagnostics14192157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Trefoil factor 1 (TFF1) plays a role in the mucus barrier. Methods: To evaluate the prevalence of TFF1 expression in cancer, a tissue microarray containing 18,878 samples from 149 tumor types and 608 samples of 76 normal tissue types was analyzed through immunohistochemistry (IHC). Results: TFF1 staining was detectable in 65 of 149 tumor categories. The highest rates of TFF1 positivity were found in mucinous ovarian carcinomas (76.2%), colorectal adenomas and adenocarcinomas (47.1-75%), breast neoplasms (up to 72.9%), bilio-pancreatic adenocarcinomas (42.1-62.5%), gastro-esophageal adenocarcinomas (40.4-50.0%), neuroendocrine neoplasms (up to 45.5%), cervical adenocarcinomas (39.1%), and urothelial neoplasms (up to 24.3%). High TFF1 expression was related to a low grade of malignancy in non-invasive urothelial carcinomas of the bladder (p = 0.0225), low grade of malignancy (p = 0.0003), estrogen and progesterone receptor expression (p < 0.0001), non-triple negativity (p = 0.0005) in invasive breast cancer of no special type, and right-sided tumor location (p = 0.0021) in colorectal adenocarcinomas. Conclusions: TFF1 IHC has only limited utility for the discrimination of different tumor entities given its expression in many tumor entities. The link between TFF1 expression and parameters of malignancy argues for a relevant biological role of TFF1 in cancer. TFF1 may represent a suitable therapeutic target due to its expression in only a few normal cell types.
Collapse
Affiliation(s)
- Florian Lutz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Soo-Young Han
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Seyma Büyücek
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Katharina Möller
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Florian Viehweger
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Ria Schlichter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Anne Menz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Andreas M. Luebke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Ahmed Abdulwahab Bawahab
- Department of Basic Medical Sciences, College of Medicine, University of Jeddah, Jeddah 21589, Saudi Arabia;
| | - Viktor Reiswich
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Sören Weidemann
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - David Dum
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
- Institute of Pathology, Clinical Center Osnabrueck, 49078 Osnabrueck, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Andreas H. Marx
- Department of Pathology, Academic Hospital Fuerth, 90766 Fuerth, Germany;
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Till Krech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
- Institute of Pathology, Clinical Center Osnabrueck, 49078 Osnabrueck, Germany
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
- Institute of Pathology, Clinical Center Osnabrueck, 49078 Osnabrueck, Germany
| | - Natalia Gorbokon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Till S. Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| |
Collapse
|
2
|
Haase A, Alefeld E, Yalinci F, Meenen DV, Busch MA, Dünker N. Gastric Inhibitory Polypeptide Receptor (GIPR) Overexpression Reduces the Tumorigenic Potential of Retinoblastoma Cells. Cancers (Basel) 2024; 16:1656. [PMID: 38730608 PMCID: PMC11083251 DOI: 10.3390/cancers16091656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Retinoblastoma (RB) is the most common malignant intraocular tumor in early childhood. Gene expression profiling revealed that the gastric inhibitory polypeptide receptor (GIPR) is upregulated following trefoil factor family peptide 1 (TFF1) overexpression in RB cells. In the study presented, we found this G protein-coupled transmembrane receptor to be co-expressed with TFF1, a new diagnostic and prognostic RB biomarker for advanced subtype 2 RBs. Functional analyses in two RB cell lines revealed a significant reduction in cell viability and growth and a concomitant increase in apoptosis following stable, lentiviral GIPR overexpression, matching the effects seen after TFF1 overexpression. In chicken chorioallantoic membrane (CAM) assays, GIPR-overexpressing RB cells developed significantly smaller CAM tumors. The effect of GIPR overexpression in RB cells was reversed by the GIPR inhibitor MK0893. The administration of recombinant TFF1 did not augment GIPR overexpression effects, suggesting that GIPR does not serve as a TFF1 receptor. Investigations of potential GIPR up- and downstream mediators suggest the involvement of miR-542-5p and p53 in GIPR signaling. Our results indicate a tumor suppressor role of GIPR in RB, suggesting its pathway as a new potential target for future retinoblastoma therapy.
Collapse
|
3
|
Hoffmann W. Self-Renewal and Cancers of the Gastric Epithelium: An Update and the Role of the Lectin TFF1 as an Antral Tumor Suppressor. Int J Mol Sci 2022; 23:ijms23105377. [PMID: 35628183 PMCID: PMC9141172 DOI: 10.3390/ijms23105377] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
In 2020, gastric cancer was the fourth leading cause of cancer deaths globally. About 90% of gastric cancers are sporadic and the vast majority are correlated with Helicobacter pylori infection; whereas familial clustering is observed in about 10% of cases. Gastric cancer is now considered to be a disease originating from dysregulated self-renewal of the gastric glands in the setting of an inflammatory environment. The human stomach contains two types of gastric units, which show bi-directional self-renewal from a complex variety of stem cells. This review focuses on recent progress concerning the characterization of the different stem cell populations and the mainly mesenchymal signals triggering their stepwise differentiation as well as the genesis of pre-cancerous lesions and carcinogenesis. Furthermore, a model is presented (Lectin-triggered Receptor Blocking Hypothesis) explaining the role of the lectin TFF1 as an antral tumor suppressor possibly regulating Lgr5+ antral stem cells in a paracrine or maybe autocrine fashion, with neighboring antral gland cells having a role as niche cells.
Collapse
Affiliation(s)
- Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| |
Collapse
|
4
|
Hwang SJ, Yeo D, Song YS, Choi Y, Youn HJ, Lee HJ. An aqueous extract from Artemisia capillaris inhibits acute gastric injury through mucosal stabilization. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:1255-1262. [PMID: 34358346 DOI: 10.1002/jsfa.11463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/19/2021] [Accepted: 08/06/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Artemisia capillaris is among the most abundantly used traditional medicines, utilized in East Asia to treat diverse illnesses, including gastrointestinal tract diseases. We previously reported that an aqueous extract of A. capillaris (AEAC) inhibited gastric inflammation induced by HCl/ethanol via reactive oxygen species scavenging and NF-κB downregulation. To date, the pharmacological potential of AEAC for promoting mucosal integrity has not been studied. RESULTS Here, we report that a single treatment with AEAC increased mucus production, and repeated administration of AEAC abolished HCl/ethanol-induced mucosal injury in vivo. Single- and multiple-dose AEAC treatments measurably increased the expression of mucosal stabilizing factors in vivo, including mucin (MUC) 5 AC, MUC6, and trefoil factor (TFF) 1 and TFF2 (but not TFF3). AEAC also induced mucosal stabilizing factors in both SNU-601 cells and RGM cells through phosphorylation of extracellular signal-regulated kinases. CONCLUSION Taken together, our results suggest that AEAC protects against HCl/ethanol-induced gastritis by upregulating MUCs and TFFs and stabilizing the mucosal epithelium. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Su Jung Hwang
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Dahee Yeo
- College of Pharmacy, Inje University, Gimhae, South Korea
| | - Ye-Seul Song
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Youngbin Choi
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Hyun-Joo Youn
- College of Pharmacy, Inje University, Gimhae, South Korea
| | - Hyo-Jong Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
5
|
Zan X, Chen Z, Guo Q, Wang Y, Zhang Z, Ji R, Zheng Y, Zhang J, Wu Z, Li M, Wang X, Ye Y, Li X, An F, Xu C, Lu L, Fan P, Zhang J, Guan Q, Li Q, Liu M, Ren Q, Hu X, Lu H, Wang Y, Zhang H, Zhao Y, Gou X, Shu X, Wang J, Hu Z, Liu R, Yuan H, Liu J, Qiao L, Zhou Y. The Association of Trefoil Factors with Gastric Cancer and Premalignant Lesions: A Cross-sectional Population-based Cohort Study. Cancer Epidemiol Biomarkers Prev 2022; 31:625-632. [PMID: 35027436 DOI: 10.1158/1055-9965.epi-21-0760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/18/2021] [Accepted: 12/22/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND A lack of research on the association of trefoil factors (TFFs) with gastric cancer (GC) and premalignant lesions (PMLs) in the general populations is an important obstacle to the application of TFFs for GC screening. We aimed to analyze the association of TFFs with GC and PMLs in a general population. METHODS We evaluated 3,986 adults residing in Wuwei, China. We collected baseline characteristics and GC risk factors, including TFFs, endoscopic diagnosis, and pathological information. Three logistic regression models were generated to analyze the association between TFFs and GC, as well as PMLs. Adjusted odds ratio (OR) and 95% confidence intervals (95% CI) were calculated to determine the strength of association. RESULTS Compared with pepsinogen (PG) and anti-Helicobacter pylori immunoglobulin G antibody (Hp-IgG), TFFs had significant association with GC and PMLs after adjusting for biomarkers and risk factors (P < 0.05). The ORs [95% CI] for TFF1 (1.67 [1.27-2.20]), TFF2 (2.66 [2.01-3.51]), and TFF3 (1.32 [1.00-1.74]) were larger than the ORs for PGI (0.79 [0.61-1.03]), PGI/II (1.00 [0.76-1.31]) and Hp-IgG (0.99 [0.73-1.35]) in the GC group. In intestinal metaplasia (IM) group, not only the TFF3 serum level was the highest, but also the OR (1.92 [1.64-2.25]) was the highest. CONCLUSIONS Trefoil factor were associated with risk of GC and PMLs. IMPACT Serum TFFs can improve the screening of high-risk populations for GC.
Collapse
Affiliation(s)
- Xiangyi Zan
- Department of Gastroenterology, The First Affiliated Hospital of Lanzhou University
| | - Zhaofeng Chen
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Qinghong Guo
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Yuping Wang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Zhiyi Zhang
- Department of Gastroenterology, Gansu Wuwei Tumor Hospital
| | - Rui Ji
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Ya Zheng
- Department of Gastroenterology, First Hospital of Lanzhou University
| | | | | | - Min Li
- School of Basic Medical Sciences, Lanzhou University
| | - Xiang Wang
- Department of Gastroenterology, Lanzhou University Second Hospital
| | - Yuwei Ye
- Department of Gastroenterology, First Hospital of Lanzhou University
| | | | | | | | | | | | - Jun Zhang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Quanlin Guan
- First Hospital of Lanzhou University, Lanzhou University
| | - Qiang Li
- The First Affiliated Hospital of Lanzhou University
| | - Min Liu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Qian Ren
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Xiaobin Hu
- Lanzhou University School of Public Health
| | - Hong Lu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Yuling Wang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Hongling Zhang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Yue Zhao
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Xi Gou
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Xiaochuang Shu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Jun Wang
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Zenan Hu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Rong Liu
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Hao Yuan
- Department of Gastroenterology, First Hospital of Lanzhou University
| | - Jiankang Liu
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School
| | - Liang Qiao
- Storr Liver Centre, the Westmead Institute for Medical Research, the University of Sydney at the Westmead
| | - Yongning Zhou
- Department of Gastroenterology, First Hospital of Lanzhou University
| |
Collapse
|
6
|
Lee DY, Song MY, Kim EH. Trefoil Factor 1 Suppresses Epithelial-mesenchymal Transition through Inhibition of TGF-beta Signaling in Gastric Cancer Cells. J Cancer Prev 2021; 26:137-144. [PMID: 34258252 PMCID: PMC8249209 DOI: 10.15430/jcp.2021.26.2.137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 01/22/2023] Open
Abstract
Gastric cancer is a malignancy with high incidence and mortality worldwide. In gastric cancer, epithelial-mesenchymal transition (EMT) and metastasis further increase the mortality rate. Trefoil factor 1 (TFF1) has been reported as a protective factor in the gastric mucosa. In this study, TFF1 inhibited the migration and invasive capability of gastric cancer cells. Elevated TFF1 levels induced the expression of E-cadherin, the epithelial marker, and reduced the expression of N-cadherin, vimentin, Snail, Twist, Zinc finger E-box binding homeobox (ZEB) 1 and ZEB2, well-known repressors of E-cadherin expression. In addition, the expression of matrix metalloproteinase (MMP)-2, MMP-7 and MMP-9, which are major markers of cancer metastasis, was suppressed by TFF1. Upregulation of TFF1 inhibited TGF-β, a major signaling for EMT induction, and the phosphorylation of Smad2/3 activated by TGF-β in AGS cells. In conclusion, TFF1 inhibits EMT through suppression of TGF-β signaling in AGS cells, which might be used in therapeutic strategies for reducing metastatic potential and invasiveness of these cells.
Collapse
Affiliation(s)
- Da-Young Lee
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, Korea
| | - Moon-Young Song
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, Korea
| | - Eun-Hee Kim
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, Korea
| |
Collapse
|
7
|
He H, Feng M, Xu H, Li X, He Y, Qin H, Zhang Y, Tang H, Zou K. Total triterpenoids from the fruits of Chaenomeles speciosa exerted gastroprotective activities on indomethacin-induced gastric damage via modulating microRNA-423-5p-mediated TFF/NAG-1 and apoptotic pathways. Food Funct 2020; 11:662-679. [PMID: 31895380 DOI: 10.1039/c9fo02322d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Our previous studies have demonstrated that the total triterpenes from the fruits of Chaenomeles speciosa (CSTT) exhibit effective therapeutic effects on gastric ulcer patients and animals. The present aim is to further investigate the mechanisms involved. The results indicated that CSTT could ameliorate IND-induced gastric injury, which was related to promoting IND-damaged GES-1 cell proliferation and migration, improving the IND-damaged rat GBF, ulcer area, inhibition rate and pathologic changes of gastric mucous tissue, increasing the amount of adhered gastric mucus, attenuating the volume and total acidity of the gastric effluents, and augmenting the gastric pH; further studies showed that CSTT obviously downregulated miR-423-5p mRNA, NAG-1 mRNA and protein expression, Bax, Bad, cytosol cytochrome C, Apaf-1, cleaved-caspase-3, and cleaved-caspase-9 protein expression and cytosol cytochrome C concentration, and upregulated TFF1, TFF2 and TFF3 mRNA and protein expression, Bcl-2, Bcl-xl, pro-caspase-3, and pro-caspase-9 protein expression, mitochondrial viability, mitochondrial cytochrome C concentration and Bcl-2/Bax, Bcl-xl/Bad ratios. These findings demonstrated that CSTT protected against IND-induced gastric damage by depressing miR-423-5p expression and modulating the TFF/NAG-1 pathway, which in turn restrained mitochondrion-mediated apoptosis.
Collapse
Affiliation(s)
- Haibo He
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang, Hubei 443002, China
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Gonzaga IM, Soares Lima SC, Nicolau MC, Nicolau-Neto P, da Costa NM, de Almeida Simão T, Hernandez-Vargas H, Herceg Z, Ribeiro Pinto LF. TFF1 hypermethylation and decreased expression in esophageal squamous cell carcinoma and histologically normal tumor surrounding esophageal cells. Clin Epigenetics 2017; 9:130. [PMID: 29296124 PMCID: PMC5738900 DOI: 10.1186/s13148-017-0429-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 11/29/2017] [Indexed: 12/21/2022] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is one of the 10 most incident cancer types in the world, and it is mainly associated with tobacco and alcohol consumption. ESCC mortality rates stand very close to its incidence, which is a direct consequence of a late diagnosis and an inefficient treatment. Although this scenery is quite alarming, the major molecular alterations that drive this carcinogenesis process remain unclear. We have previously shown through the first ESCC methylome analysis that TFF1 promoter is frequently hypermethylated in ESCC. Here, to evaluate TFF1 methylation as a potential biomarker of early ESCC diagnosis, we investigated the status of TFF1 promoter methylation and its expression in ESSC and histologically normal tumor surrounding tissue of ESCC patients in comparison to healthy esophagus of non-cancer individuals. Results Analysis of TFF1 promoter methylation, and gene and protein expression in 65 ESCC patients and 88 controls revealed that TFF1 methylation levels were already increased in histologically normal tumor surrounding tissue of ESCC patients when compared to healthy esophagus of non-cancer individuals. This increase in DNA methylation was followed by the reduction of TFF1 mRNA expression. Interestingly, TFF1 expression was capable of distinguishing tumor surrounding normal tissue from normal mucosa of healthy individuals with 92% accuracy. In addition, TFF1 protein was undetectable both in tumor and surrounding mucosa by immunohistochemistry, while submucosa glands of the healthy esophagus showed positive staining. Furthermore, treatment of TE-1 and TE-13 ESCC cell lines with decitabine led to a reduction of promoter methylation and consequent upregulation of TFF1 gene and protein expression. Finally, using TCGA data we showed that TFF1 loss is observed in ESCC, but not in esophageal adenocarcinoma, highlighting the different molecular mechanisms involved in the development of each histological subtype of esophageal cancer. Conclusions This study shows that TFF1 expression is silenced in early phases of ESCC development, which seems to be mediated at least in part by promoter hypermethylation, and provides the basis for the use of TFF1 expression as a potential biomarker for early ESCC detection.
Collapse
Affiliation(s)
- Isabela Martins Gonzaga
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer, Coordenação de Pesquisa, Rua André Cavalcanti, 37–6° andar, Bairro de Fátima, Rio de Janeiro, Rio de Janeiro CEP: 20231-050 Brazil
| | - Sheila Coelho Soares Lima
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer, Coordenação de Pesquisa, Rua André Cavalcanti, 37–6° andar, Bairro de Fátima, Rio de Janeiro, Rio de Janeiro CEP: 20231-050 Brazil
| | - Marina Chianello Nicolau
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer, Coordenação de Pesquisa, Rua André Cavalcanti, 37–6° andar, Bairro de Fátima, Rio de Janeiro, Rio de Janeiro CEP: 20231-050 Brazil
| | - Pedro Nicolau-Neto
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer, Coordenação de Pesquisa, Rua André Cavalcanti, 37–6° andar, Bairro de Fátima, Rio de Janeiro, Rio de Janeiro CEP: 20231-050 Brazil
| | - Nathalia Meireles da Costa
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer, Coordenação de Pesquisa, Rua André Cavalcanti, 37–6° andar, Bairro de Fátima, Rio de Janeiro, Rio de Janeiro CEP: 20231-050 Brazil
| | - Tatiana de Almeida Simão
- Departamento de Bioquímica, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Av. 28 de Setembro 87 fundos, Vila Isabel, Rio de Janeiro, CEP: 20551-013 Brazil
| | - Hector Hernandez-Vargas
- Epigenetics Group, Section of Mechanisms of Carcinogenesis, International Agency for Research on Cancer, 150 Cours Albert Thomas, 69372, CEDEX 08 Lyon, France
| | - Zdenko Herceg
- Epigenetics Group, Section of Mechanisms of Carcinogenesis, International Agency for Research on Cancer, 150 Cours Albert Thomas, 69372, CEDEX 08 Lyon, France
| | - Luis Felipe Ribeiro Pinto
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer, Coordenação de Pesquisa, Rua André Cavalcanti, 37–6° andar, Bairro de Fátima, Rio de Janeiro, Rio de Janeiro CEP: 20231-050 Brazil
- Departamento de Bioquímica, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Av. 28 de Setembro 87 fundos, Vila Isabel, Rio de Janeiro, CEP: 20551-013 Brazil
| |
Collapse
|
9
|
Yusup A, Huji B, Fang C, Wang F, Dadihan T, Wang HJ, Upur H. Expression of trefoil factors and TWIST1 in colorectal cancer and their correlation with metastatic potential and prognosis. World J Gastroenterol 2017; 23:110-120. [PMID: 28104986 PMCID: PMC5221274 DOI: 10.3748/wjg.v23.i1.110] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/19/2016] [Accepted: 12/02/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To detect the expression of trefoil factors (TFFs) and TWIST1 in colorectal cancer (CRC) and analyze their correlation with metastasis and survival.
METHODS This study examined the expression of TFF1, TFF3 and TWIST1 in a total of 75 tumor samples, 47 matched normal samples (15 cm from the lesion margin), 30 metastatic lymph nodes, and 10 liver metastatic cancer samples from patients with CRC. The relationship was then analyzed between the protein expression and different clinical records. TFF1, TFF3, TWIST1,E-cadherin, vimentin and β-catenin mRNA and protein expression levels were measured in colon cancer cell lines with different metastatic potentials (HIEC, HT29, SW620, and LoVo cells), and the correlation of the expression levels with epithelial-mesenchymal transition (EMT) was discussed.
RESULTS It was found that 66.7% (50/75), 78.7% (59/75) and 54.7% (41/75) of tumor tissue samples exhibited positive staining for TFF1, TFF3 and TWIST1 and so did 27.3% (13/47), 100% (47/47) and 17% (8/47) of adjacent normal colorectal tissues. Compared with adjacent normal tissues, significant differences were found in the expression of all three proteins in different cancerous tissues (P < 0.05). Higher expression of TFF3 and TWIST1 was significantly correlated with lymph node metastasis (P = 0.034, P = 0.000), advanced stage (P = 0.031, P = 0.003), and poorer survival (P = 0.042 for the TFF3 group, P = 0.003 for the TWIST1 group). The expression of TFF3 and TWIST1 in cancer cell lines was higher than that in HIEC (a normal human intestinal epithelial cell line)(P < 0.05), and the expression intensity demonstrated a tendency to rise with increased metastatic potential both at the protein and mRNA levels. However, TFF1 expression demonstrated the opposite tendency. It was also observed that the expression of E-cadherin and β-catenin tended to decrease while that of vimentin, TWIST1 and Snail tended to rise with the increase in metastatic potential.
CONCLUSION The expression of TFF3 and TWIST1 might be associated with the survival of patients with CRC after curative resection and might be pivotal predictors of disease progression. TFF3 may be correlated to the invasiveness of CRC.
Collapse
|
10
|
Song X, Xin N, Wang W, Zhao C. Wnt/β-catenin, an oncogenic pathway targeted by H. pylori in gastric carcinogenesis. Oncotarget 2016; 6:35579-88. [PMID: 26417932 PMCID: PMC4742126 DOI: 10.18632/oncotarget.5758] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/26/2015] [Indexed: 12/14/2022] Open
Abstract
A section of gastric cancers presents nuclear β-catenin accumulation correlated with H. pylori infection. H. pylori stimulate Wnt/β-catenin pathway by activating oncogenic c-Met and epidermal growth factor receptor (EGFR), or by inhibiting tumor suppressor Runx3 and Trefoil factor 1 (TFF1). H. pylori also trigger Wnt/β-catenin pathway by recruiting macrophages. Moreover, Wnt/β-catenin pathway is found involved in H. pylori-induced gastric cancer stem cell generation. Recently, by using gastroids, researchers have further revealed that H. pylori induce gastric epithelial cell proliferation through β-catenin. These findings indicate that Wnt/β-catenin is an oncogenic pathway activated by H. pylori. Therefore, this pathway is a potential therapy target for H. pylori-related gastric cancer.
Collapse
Affiliation(s)
- Xiaowen Song
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Na Xin
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Wei Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Chenghai Zhao
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
11
|
Busch M, Dünker N. Trefoil factor family peptides – friends or foes? Biomol Concepts 2015; 6:343-59. [DOI: 10.1515/bmc-2015-0020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 08/20/2015] [Indexed: 12/13/2022] Open
Abstract
AbstractTrefoil 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.
Collapse
Affiliation(s)
- Maike Busch
- 1Medical Faculty, Institute for Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, Hufelandstr. 55, D-45122 Essen, Germany
| | - Nicole Dünker
- 1Medical Faculty, Institute for Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, Hufelandstr. 55, D-45122 Essen, Germany
| |
Collapse
|
12
|
Xiao P, Ling H, Lan G, Liu J, Hu H, Yang R. Trefoil factors: Gastrointestinal-specific proteins associated with gastric cancer. Clin Chim Acta 2015; 450:127-34. [PMID: 26265233 DOI: 10.1016/j.cca.2015.08.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 08/07/2015] [Accepted: 08/07/2015] [Indexed: 12/11/2022]
Abstract
Trefoil factor family (TFF), composed of TFF1, TFF2, and TFF3, is a cluster of secreted peptides characterized by trefoil domain (s) and C-terminal dimerization domain. TFF1, a gastric tumor suppressor, is a single trefoil peptide originally detected in breast cancer cell lines but expressed mainly in the stomach; TFF2, a candidate of gastric cancer suppressor with two trefoil domains, is abundant in the stomach and duodenal Brunner's glands; and TFF3 is another single trefoil peptide expressed throughout the intestine which can promote the development of gastric carcinoma. According to multiple studies, TFFs play a regulatory function in the mammals' digestive system, namely in mucosal protection and epithelial cell reconstruction, tumor suppression or promotion, signal transduction and the regulation of proliferation and apoptosis. Action mechanisms of TFFs remain unresolved, but the recent demonstration of a GKN (gastrokine) 2-TFF1 heterodimer implicates structural and functional interplay with gastrokines. This review aims to encapsulate the structural and biological characteristics of TFF.
Collapse
Affiliation(s)
- Ping Xiao
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| | - Hui Ling
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China.
| | - Gang Lan
- Key Laboratory for Atherosclerology of Hunan Province, Cardiovascular Research Institute, University of South China, Hengyang, Hunan 421001, PR China
| | - Jiao Liu
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| | - Haobin Hu
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| | - Ruirui Yang
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| |
Collapse
|
13
|
Huang Z, Zhang X, Lu H, Wu L, Wang D, Zhang Q, Ding H. Serum trefoil factor 3 is a promising non-invasive biomarker for gastric cancer screening: a monocentric cohort study in China. BMC Gastroenterol 2014; 14:74. [PMID: 24720760 PMCID: PMC4012276 DOI: 10.1186/1471-230x-14-74] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 03/24/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The search for better non-invasive biomarkers for gastric cancer remains ongoing. We investigated the predictive power of serum trefoil factor (TFF) levels as biomarkers for gastric cancer in comparison with the pepsinogen (PG) test. METHODS Patients with gastric cancer, chronic atrophic gastritis (CAG) or chronic non-atrophic gastritis (CNAG), and healthy people were recruited. Serum concentrations of TFFs, PG I, and PG II, as well as the presence of antibodies against Helicobacter pylori, were measured by enzyme-linked immunosorbent assays (ELISA). Receiver operating characteristics (ROC) were used to compare the predictive powers of the selected factors. RESULTS The serum concentrations of TFF1, TFF2, and TFF3 in the control groups were significantly lower than those in the gastric cancer group with the exception of TFF2 which was elevated in CAG. The area under the ROC curve for TFF3 was greater than that for the PG I/II ratio (0.81 vs 0.78). TFF3 also had a significantly higher predictive power for distinguishing gastric cancer than the PG test (odds ratio: 10.33 vs 2.57). Moreover, combining the serum TFF3 and PG tests for gastric cancer had better predictive power than either alone. CONCLUSIONS Serum TFF3 may be a better predictor of gastric cancer than the PG test, while the combined testing of serum PG and TFF3 could further improve the efficacy of gastric cancer screening.
Collapse
Affiliation(s)
- Zhigang Huang
- Department of Gastroenterology, Lihuili Hospital of Ningbo Medical Center, 57# Xingning Road, Ningbo 315000, China.
| | | | | | | | | | | | | |
Collapse
|
14
|
Copper promotes TFF1-mediated Helicobacter pylori colonization. PLoS One 2013; 8:e79455. [PMID: 24236136 PMCID: PMC3827375 DOI: 10.1371/journal.pone.0079455] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 10/01/2013] [Indexed: 02/08/2023] Open
Abstract
The trefoil peptides (TFF1, TFF2 and TFF3) are a family of small highly conserved proteins that play an essential role in epithelial regeneration within the gastrointestinal tract, where they are mainly expressed. TFF1 expression is strongly induced after mucosal injury and it has been proposed that tff1 functions as a gastric tumor suppressor gene. Several studies confirm that tff1 expression is frequently lost in gastric cancer because of deletions, mutations or methylation of the tff1 promoter. Infection by Helicobacter pylori (H. pylori) results in chronic gastritis and it can lead to the development of gastric or duodenal ulcers. Moreover, it is known that there is a strong link to the development of gastric cancer. It has been shown that H. pylori interacts with the dimeric form of TFF1 and that the rough form of lipopolysaccharide mediates this interaction. We have previously reported that the carboxy-terminus of TFF1 is able to specifically bind copper ions (Cu) and that Cu binding favours the homodimerization of the peptide, thus enhancing its motogenic activity. Here, we report that the Cu-TFF1 cuprocomplex promotes adherence of H. pylori to epithelial cells. Adherence of H. pylori to gastric adenocarcinoma cells, AGS AC1 cells, induced to hyper-express TFF1 was enhanced compared to noninduced cells. Copper further promoted this interaction. A H. pylori mutant unable to bind TFF1 did not show enhanced infection of induced cells. Cu treatment induced a thickening of the mucus layer produced by the colorectal adenocarcinoma mucus secreting, goblet cells, HT29-E12 and promoted H. pylori colonisation. Finally, SPR analysis shows that the C-terminus of TFF1, involved in the binding of copper, is also able to selectively bind H. pylori RF-LPS.
Collapse
|
15
|
Huang YG, Li YF, Pan BL, Wang LP, Zhang Y, Lee WH, Zhang Y. Trefoil factor 1 gene alternations and expression in colorectal carcinomas. TUMORI JOURNAL 2013; 99:702-7. [DOI: 10.1177/030089161309900610] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Aims and backgroundAberrant expression of the trefoil factor family (TFF) has been recognized to be involved in the development and/or progression of various solid tumors. Increased trefoil factor 1 (TFF1) expression is found associated with tumor progression in some tumors, and TFF1 missense mutations have been detected in gastric cancer. The aim of the study was to analyze TFF1 alternations and expression in colorectal carcinoma and their correlation with cancer progression and pathological aspects.MethodsTFF1 mutations were detected in colorectal carcinomas by DNA sequencing. TFF1mRNA and protein levels in subsets of the primary tumors were determined using quantitative reverse transcription polymerase chain reaction and immunohistochemistry analyses. The serum level of TFF1 was also detected by enzyme-linked immunosorbent assay for patients with colorectal carcinoma.ResultsFive variants were detected in the 5'-untranslation region and intron 1 of TFF1. TFF1 expression was increased in colorectal carcinoma compared to paired distal colonic mucosa. Immunohistochemistry in primary colorectal carcinoma showed no significant differences in tumor TFF1 levels with respect to clinicopathological parameters such as the patient's sex, cancer differentiation, stage and lymph node metastasis. However, serum TFF1 levels were significantly elevated in patients with colorectal carcinoma compared to healthy individuals.ConclusionsThe results indicate that TFF1 missense mutations seem to be a rare event in colorectal carcinogenesis. Serum TFF1 may be a potential useful marker for patients with colorectal carcinoma.
Collapse
Affiliation(s)
- You-Guang Huang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology
- Tumor institue of Yunnan Province, The 3rd Affiliated Hospital of Kunming
| | - Yun-Feng Li
- Tumor institue of Yunnan Province, The 3rd Affiliated Hospital of Kunming
| | - Bao-Long Pan
- Department of Clinical Laboratory, The 1st Hospital of Yuxi, Yunnan Province
| | - Li-Ping Wang
- Department of Pathology, Yan-an Hospital of Kunming, Kunming, China
| | - Yong Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology
| | - Wen-Hui Lee
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology
| | - Yun Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology
| |
Collapse
|
16
|
Tanaka T, Nakamura J, Kitajima Y, Kai K, Miyake S, Hiraki M, Ide T, Koga Y, Noshiro H. Loss of trefoil factor 1 is regulated by DNA methylation and is an independent predictive factor for poor survival in advanced gastric cancer. Int J Oncol 2013; 42:894-902. [PMID: 23291975 DOI: 10.3892/ijo.2013.1759] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 11/30/2012] [Indexed: 02/05/2023] Open
Abstract
Trefoil factor 1 (TFF1) is considered to be a tumor suppressor gene in gastric cancer. However, the role of TFF1 expression and its regulation in gastric cancer patients remain unclear. The aims of this study were to clarify the clinical significance of TFF1 and to determine its regulatory mechanisms. We assessed the immunohistochemical expression of TFF1 in 182 gastric cancer patients and examined whether or not TFF1 is associated with the clinicopathological factors and patient survival. In vitro study using TFF1 knockdown gastric cancer cells evaluated the role of TFF1 in cancer invasion. Bisulfite sequencing was performed to assess DNA methylation of TFF1 in cells and resected tissues. Patients with low expression of TFF1 showed a significantly deeper invasion of the tumor than those with high expression (p=0.037). Low expression of TFF1 was also associated with a poor survival (p=0.029) in 108 patients who were treated by surgery alone. Both TFF1 expression and lymph node metastasis are independent predictive factors for disease-specific survival in a multivariate analysis. In an in vitro study, invasive power of the cells was significantly increased in the TFF1‑deficient cells compared with the control cells. Bisulfate sequencing showed that TFF1 expression is strongly dependent on DNA methylation in both gastric cancer cells and tissues. Interestingly, methylation status of two specific CpG sites, which are located close to a TATA box and hypoxia response element (HRE), determined the TFF1 expression in the resected tissues. TFF1 expression is silenced by DNA methylation and is associated with tumor invasion and a poor survival in gastric cancer patients. The expression and̸or methylation status of TFF1 may, therefore, serve as a useful biomarker for predicting survival in patients with advanced gastric cancer.
Collapse
Affiliation(s)
- Tomokazu Tanaka
- Department of Surgery, Saga University Faculty of Medicine, Saga 849-8501, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Ge Y, Zhang J, Cao J, Wu Q, Sun L, Guo L, Wang Z. TFF1 inhibits proliferation and induces apoptosis of gastric cancer cells in vitro. Bosn J Basic Med Sci 2012; 12:74-81. [PMID: 22642590 DOI: 10.17305/bjbms.2012.2499] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Trefoil Factor Family (TFF) plays an essential role in the intestinal epithelial restitution, but the relationship between TFF1 and gastric cancer (GC) is still unclear. The present study aimed to determine the role of TFF1 in repairing gastric mucosa and in the pathogenesis of GC. The TFF1 expression in different gastric mucosas was measured with immunohistochemistry. Then, siRNA targeting TFF1 or plasmids expressing TFF1 gene were transfected into BGC823 cells, SGC7901 cells and GES-1 cells. The cell proliferation was detected with MTT assay and apoptosis and cell cycle measured by flow cytometry. From normal gastric mucosa to mucosa with dysplasia and to gastric cancer, the TFF1 expression had a decreasing trend. Down-regulation of TFF1 expression significantly reduced the apoptosis of three cell lines and markedly facilitated their proliferation but had no significant effect on cell cycle. Over-expression of TFF1 could promote apoptosis of three cell lines and inhibit proliferation but had no pronounced effect on cell cycle. TFF1 can inhibit proliferation and induce apoptosis of GC cells in vitro.
Collapse
Affiliation(s)
- Yanli Ge
- Department of Gastroenterology, Tongji Hospital, Tongji University, Shanghai 200065, China
| | | | | | | | | | | | | |
Collapse
|
18
|
Estrogen is increased in male cholangiocarcinoma patients' serum and stimulates invasion in cholangiocarcinoma cell lines in vitro. J Cancer Res Clin Oncol 2012; 138:1311-20. [PMID: 22476540 DOI: 10.1007/s00432-012-1207-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 03/19/2012] [Indexed: 02/07/2023]
Abstract
PURPOSE Cholangiocarcinoma is defined as a chronic liver disease with altered estrogen metabolism and could result in estrogen retention. Estrogenic response was known as a promoting factor in progression of some cancer. In this study, we determined the significant increase of estrogen level in cholangiocarcinoma patients' sera. METHODS The estrogen levels in cholangiocarcinoma patients' sera were measured and correlated with clinical presentations. Estrogen receptor-α expressions in cholangiocarcinoma tissues were detected by immunohistochemistry method. KKU-100 and KKU-M213 cholangiocarcinoma cell lines were treated with 17β-estradiol and tested the proliferative and invasive effects. RESULTS The estrogen levels showed positive correlations with serum bilirubin and alkaline phosphatase and a negative correlation with albumin. This study also showed an association with shorter survival times when patients with low and high serum estrogen levels were compared. In vitro studies demonstrated the effect of estrogen on cell proliferation and invasion in dose-dependent manners, which could be inhibited by tamoxifen, a clinical used estrogen antagonist. Invasion showed an association with the TFF1 gene expression and could be inhibited by small interfering RNA against TFF1 gene. Estrogen receptor-α was the main estrogen receptor that response to 17β-estradiol stimulation. CONCLUSIONS TFF1 trefoil protein could be one of the effectors for estrogen-induced invasion in cholangiocarcinoma via the estrogen receptor-α. These findings could lead to an understanding of the mechanism of cholangiocarcinoma progression.
Collapse
|
19
|
Qu Y, Yang Y, Ma D, Xiao W. Increased trefoil factor 3 levels in the serum of patients with three major histological subtypes of lung cancer. Oncol Rep 2012; 27:1277-83. [PMID: 22246423 PMCID: PMC3583529 DOI: 10.3892/or.2012.1627] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 12/22/2011] [Indexed: 01/11/2023] Open
Abstract
Lung cancer is the most common cause of cancer-related deaths in the world. The trefoil factor (TFF) family is composed of three thermostable, and protease-resistant proteins, named TFF1, TFF2 and TFF3. TFF protein levels have been found to be related to the development of various types of cancer. However, it is still unclear whether TFF proteins are differentially expressed in the serum of different histological subtypes of lung cancer compared to healthy individuals. In this study, we investigated the levels of TFF proteins in serum and lung tissues of 130 lung cancer patients (58 squamous cell lung carcinoma cases, 43 adenocarcinoma cases and 29 SCLC cases) and 60 healthy individuals. It was found that TFF1 and TFF2 have similar or slightly higher levels in these three subtypes of lung cancer compared to healthy individuals, while TFF3 levels were significantly higher in the examined lung cancer cases compared to healthy individuals. Immunoblot analyses of TFF1, TFF2 and TFF3 indicated that lung cancer tissues and lung cancer cell lines have a higher expression of the TFF3 protein, but not of TFF1 or TFF2 proteins, compared to tissues from healthy individuals or from the normal cell line. Quantitative RT-PCR analysis indicated higher levels of TFF3, but not TFF1 and TFF2, transcripts in lung cancer tissues or cell lines. These results show increased TFF3 levels in serum and lung tissues, suggesting that TFF3 may serve as a promising, easily detected biomarker of lung cancer.
Collapse
Affiliation(s)
- Yiqing Qu
- Department of Respiratory Medicine, Qilu Hospital, Shandong University, Jinan 250012, PR China.
| | | | | | | |
Collapse
|
20
|
Zhao L, Yang J, Yu J, Zhang H, Gao DL, Chen KS. Screening of genes differentially expressed between human esophageal squamous cell carcinoma and tumor-adjacent normal tissue using gene chip technology. Shijie Huaren Xiaohua Zazhi 2011; 19:2328-2333. [DOI: 10.11569/wcjd.v19.i22.2328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To screen differentially expressed genes between human esophageal squamous cell carcinoma and tumor-adjacent normal tissue to lay a theoretical basis for finding molecular markers specific for esophageal squamous cell carcinoma.
METHODS: Total RNA was extracted from human esophageal squamous cell carcinoma and tumor adjacent normal tissue specimens and cDNA was synthesized by reverse transcription. Probes were prepared by labeling the synthesized cDNA with Cy3-dUTP and hybridized with gene chips to screen differentially expressed genes. Bioinformatic tools were used for characterization of differentially expressed genes obtained.
RESULTS: According to the expression ratio of >4.0 or <0.25, a total of 1 113 differentially expressed genes were identified, including 464 up-regulated and 649 down-regulated ones. Bioinformatic analysis identified a variety of known functional genes. RT-PCR analysis confirmed that three genes were differentially expressed between human esophageal squamous cell carcinoma and tumor adjacent normal tissue.
CONCLUSION: Gene chip technology is highly efficient and reliable in screening genes differentially expressed in esophageal squamous cell carcinoma. The occurrence and development of esophageal squamous cell carcinoma is the result of expression of many cancer-related genes.
Collapse
|
21
|
Affiliation(s)
- Won Sang Park
- Department of Pathology, The Catholic University of Korea, School of Medicine,
| |
Collapse
|
22
|
Bornschein J, Weigt J, Selgrad M, Malfertheiner P. Molecular aspects in the diagnosis of gastric cancer. ACTA ACUST UNITED AC 2009; 3:585-96. [PMID: 23495987 DOI: 10.1517/17530050902862175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Gastric cancer (GC) represents the second most common cause of cancer-related death worldwide. The prognosis remains poor, with limited treatment options. A better understanding of the initiation and progression of GC would enable the development of general screening strategies and individualized treatment modalities. OBJECTIVE The assessment of tools and molecular markers for the early detection and diagnosis of GC. METHODS Human clinical studies published within the past 5 years are reviewed. Also, significant previous data on markers in clinical use or on relevant animal or cell culture experiments are considered. RESULTS/CONCLUSION Serum-based screening strategies are not ready for routine application but represent an opportunity for the identification of individuals at high risk with the need for primary gastroscopy and further surveillance, which would ultimately improve survival and prognosis of GC. Infection with Helicobacter pylori represents the principal risk factor for gastric carcinogenesis. Bacterial virulence and host genetic factors contribute to individual susceptibility. Key molecular alterations in gastric carcinogenesis are related to intra- and extracellular cascades that regulate cell proliferation, tumor invasion and metastastic spread. For the development of effective prevention and treatment modalities, it is essential to unravel the basic mechanisms of gastric carcinogenesis.
Collapse
Affiliation(s)
- Jan Bornschein
- Otto-von-Guericke-University of Magdeburg, Department of Gastroenterology, Hepatology and Infectious Diseases, Leipziger Str. 44, D-39120 Magdeburg, Germany +0049 391 6713100 ; +0049 391 6713105 ;
| | | | | | | |
Collapse
|
23
|
Karam SM, Tomasetto C, Rio M. Amplification and invasiveness of epithelial progenitors during gastric carcinogenesis in trefoil factor 1 knockout mice. Cell Prolif 2008; 41:923-935. [PMID: 19040570 PMCID: PMC6496233 DOI: 10.1111/j.1365-2184.2008.00562.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2007] [Accepted: 03/12/2008] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE It is not known whether or not epithelial progenitors of the pyloric antrum are involved in gastric carcinogenesis. Normally, these progenitors give rise to two main cell lineages: pit and gland mucous cells. This study was designed to examine the changes that occur in pyloric antral mucous cell lineages and their progenitors during development of gastric adenoma and carcinoma in trefoil factor 1 (TFF1) knockout mice. MATERIALS AND METHODS Pyloric antral mucosal tissues of TFF1 knockout mice at ages from 3 days to 17 months were processed for histochemical analysis using Ulex europaeus and Grifforia simplifolica lectins as markers for pit and gland mucous cells, respectively. The dividing epithelial progenitors were identified by using immunohistochemical and electron microscopy techniques. RESULTS TFF1 loss was associated with amplification of both mucus-secreting pit and gland cells. Both lectins examined bound not only to mature mucous cells, but also to most of epithelial progenitors which gradually amplified with age and frequently were seen in mitosis. Analysis of 12- to 17-month-old TFF1-deficient stomachs revealed occasional groups of poorly differentiated mucosal cells with features similar to those of epithelial progenitors (or stem cells), in the basal portion of the antral mucosa. These cells eventually invaded the muscularis mucosa while maintaining some capacity to differentiate. CONCLUSION This study shows that the progenitors of pit and gland mucous cells contribute to gastric carcinogenesis in the pyloric antrum of TFF1 knockout mice, strongly supporting the concept of stem cell origin of cancer.
Collapse
Affiliation(s)
- S. M. Karam
- Department of Anatomy, Faculty of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates, and
| | - C. Tomasetto
- Institute of Genetics and Molecular and Cellular Biology, Department of Cancer Biology, University of Louis Pasteur, Illkirch cedex, France
| | - M.‐C. Rio
- Institute of Genetics and Molecular and Cellular Biology, Department of Cancer Biology, University of Louis Pasteur, Illkirch cedex, France
| |
Collapse
|
24
|
Perry JK, Kannan N, Grandison PM, Mitchell MD, Lobie PE. Are trefoil factors oncogenic? Trends Endocrinol Metab 2008; 19:74-81. [PMID: 18054496 DOI: 10.1016/j.tem.2007.10.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Revised: 10/19/2007] [Accepted: 10/22/2007] [Indexed: 12/24/2022]
Abstract
Trefoil factors (TFFs), in particular TFF1, are classical estrogen-regulated genes and have served as markers of estrogen gene regulation by various environmental estrogens. TFFs are also regulated by several other factors including growth hormone (hGH), insulin-like growth factor-1 (IGF-1), epidermal growth factor (EGF) and various oncogenic stimuli. TFFs are secreted proteins present in serum and possess the potential to act as growth factors promoting cell survival, anchorage-independent growth and motility. Recent compelling evidence has emerged from experimental and clinical studies to indicate a pivotal role of TFFs in oncogenic transformation, growth and metastatic extension of common human solid tumours. This review will summarize the current evidence for the involvement of TFFs in human cancer.
Collapse
Affiliation(s)
- Jo K Perry
- Liggins Institute, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | | | | | | |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW To summarize key aspects from recent research as well as review articles on the topic of genetic mouse models, particularly in knockout mice, that have considerably contributed to understanding the pathways and mechanisms underlying gastric physiology. RECENT FINDINGS A series of knockout mouse models has proven to be invaluable in elucidating the mechanism and validating the current model of acid secretion. The interaction between the gastrin-histamine and cholecystokinin-somatostatin pathways was identified using the genetic approach as being critical in regulating acid secretion. Curiously, neither ghrelin nor ghrelin receptor knockout mice displayed the expected lean phenotype. Importantly, the study of obestatin in GPR39 knockout mice could be misleading, as zinc rather than obestatin is the endogenous ligand for GPR39. The physiological roles of ghrelin and obestatin have yet to be confirmed using knockout mouse models. SUMMARY The knockout mouse continues to serve as an excellent model to dissect the complexity of the mechanism of gastric acid secretion and to study the physiological importance of gastric ghrelin.
Collapse
Affiliation(s)
- Duan Chen
- Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway.
| | | |
Collapse
|
26
|
Sasaki M, Ikeda H, Nakanuma Y. Expression profiles of MUC mucins and trefoil factor family (TFF) peptides in the intrahepatic biliary system: physiological distribution and pathological significance. ACTA ACUST UNITED AC 2007; 42:61-110. [PMID: 17616258 DOI: 10.1016/j.proghi.2007.02.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mucin secreted by mucosal epithelial cells plays a role in the protection of the mucosal surface and also is involved in pathological processes. So far, MUC1-4, 5AC, 5B, 6-8, 11-13 and 15-17 genes coding the backbone mucin core protein have been identified in humans. Their diverse physiological distribution and pathological alterations have been reported. Trefoil factor family (TFF) peptides are mucin-associated molecules co-expressed with MUC mucins and involved in the maintenance of mucosal barrier and the biological behavior of epithelial and carcinoma cells. Intrahepatic biliary system is a route linking the bile canaliculi and the extrahepatic bile duct for the excretion of bile synthesized by hepatocytes. Biliary epithelial cells line in the intrahepatic biliary system, secreting mucin and other molecules involved in the maintenance and regulation of the system. In this review, the latest information regarding properties, expression profiles and regulation of MUC mucins and TFF peptides in the intrahepatic biliary system is summarized. In particular, we focus on the expression profiles and their significance of MUC mucins in developmental and normal livers, various hepatobiliary diseases and intrahepatic cholangiocarcinoma.
Collapse
Affiliation(s)
- Motoko Sasaki
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Takaramachi 13-1, Kanazawa 920-8640, Japan.
| | | | | |
Collapse
|
27
|
Giraud AS, Jackson C, Menheniott TR, Judd LM. Differentiation of the Gastric Mucosa IV. Role of trefoil peptides and IL-6 cytokine family signaling in gastric homeostasis. Am J Physiol Gastrointest Liver Physiol 2007; 292:G1-5. [PMID: 16935852 DOI: 10.1152/ajpgi.00382.2006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gastric trefoil peptides mediate mucosal repair by stimulating cell migration, inhibiting apoptosis and inflammation, and likely augmenting the barrier function of mucus. One of these, tff1, is a gastric-specific tumor suppressor gene, which when repressed is associated with gastric cancer progression. IL-6 family cytokines play an important role in maintaining gastric homeostasis by regulating tff1 and other mediators of mucosal proliferation, inflammation, angiogenesis, and apoptosis. In this review the signaling cascades downstream of the common IL-6 cytokine family coreceptor gp130 that contribute to control of this homeostasis are described, as are the pathological outcomes of imbalancing these pathways.
Collapse
Affiliation(s)
- A S Giraud
- Department of Medicine, University of Melbourne at Western Hospital, Footscray, Australia.
| | | | | | | |
Collapse
|