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
|
Meng X, Liu J, Kang J, Wang M, Guan Z, Tian D, Chen X. Lamivudine protects mice from gastric ulcer by activating PGK1 to suppress ferroptosis. Biochem Pharmacol 2024; 227:116440. [PMID: 39029631 DOI: 10.1016/j.bcp.2024.116440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
Gastric ulcer is a highly prevalent digestive tract disease across the world, which is recurrent and hard to cure, sometimes transforming into gastric cancer if left untreated, posing great threat to human health. To develop new medicines for gastric ulcer, we ran a series of screens with ethanol stress model in GES-1 cells, and we uncovered that lamivudine rescued cells from ethanol toxicity. Then, we confirmed this discovery using the well-established ethanol-induced gastric ulcer model in mice and our findings suggest that lamivudine can directly activate phosphoglycerate kinase 1 (PGK1, EC 2.7.2.3), which binds and stimulates superoxide dismutase 1 (SOD1, EC 1.15.1.1) to inhibit ferroptosis and ultimately improve gastric ulcer. Moreover, AAV-PGK1 exhibited comparable gastroprotective effects to lamivudine. The findings are expected to offer novel therapeutic strategies for gastric ulcer, encompassing both lamivudine and AAV-PGK1.
Collapse
Affiliation(s)
- Xinrui Meng
- Department of Pharmacy, Lanzhou University, Lanzhou 730000, PR China; Southeast Research Institute, Lanzhou University, Putian 351152, PR China
| | - Jingjing Liu
- Department of Pharmacy, Lanzhou University, Lanzhou 730000, PR China; Southeast Research Institute, Lanzhou University, Putian 351152, PR China
| | - Jia Kang
- Department of Pharmacy, Lanzhou University, Lanzhou 730000, PR China; Southeast Research Institute, Lanzhou University, Putian 351152, PR China
| | - Menghan Wang
- Department of Pharmacy, Lanzhou University, Lanzhou 730000, PR China; Southeast Research Institute, Lanzhou University, Putian 351152, PR China
| | - Zhanghui Guan
- Department of Pharmacy, Lanzhou University, Lanzhou 730000, PR China; Southeast Research Institute, Lanzhou University, Putian 351152, PR China
| | - Dong Tian
- Department of Pharmacy, Lanzhou University, Lanzhou 730000, PR China; Southeast Research Institute, Lanzhou University, Putian 351152, PR China
| | - Xinping Chen
- Department of Pharmacy, Lanzhou University, Lanzhou 730000, PR China; Southeast Research Institute, Lanzhou University, Putian 351152, PR China.
| |
Collapse
|
3
|
Peiffer AL, Dugan AE, Kiessling LL. Soluble Human Lectins at the Host-Microbe Interface. Annu Rev Biochem 2024; 93:565-601. [PMID: 38640018 PMCID: PMC11296910 DOI: 10.1146/annurev-biochem-062917-012322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
Human lectins are integral to maintaining microbial homeostasis on the skin, in the blood, and at mucosal barriers. These proteins can recognize microbial glycans and inform the host about its microbial status. In accordance with their roles, their production can vary with tissue type. They also can have unique structural and biochemical properties, and they can influence microbial colonization at sites proximal and distal to their tissue of origin. In line with their classification as innate immune proteins, soluble lectins have long been studied in the context of acute infectious disease, but only recently have we begun to appreciate their roles in maintaining commensal microbial communities (i.e., the human microbiota). This review provides an overview of soluble lectins that operate at host-microbe interfaces, their glycan recognition properties, and their roles in physiological and pathological mechanisms.
Collapse
Affiliation(s)
- Amanda L Peiffer
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| | - A E Dugan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| | - L L Kiessling
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| |
Collapse
|
4
|
Bechtella L, Chunsheng J, Fentker K, Ertürk GR, Safferthal M, Polewski Ł, Götze M, Graeber SY, Vos GM, Struwe WB, Mall MA, Mertins P, Karlsson NG, Pagel K. Ion mobility-tandem mass spectrometry of mucin-type O-glycans. Nat Commun 2024; 15:2611. [PMID: 38521783 PMCID: PMC10960840 DOI: 10.1038/s41467-024-46825-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/12/2024] [Indexed: 03/25/2024] Open
Abstract
The dense O-glycosylation of mucins plays an important role in the defensive properties of the mucus hydrogel. Aberrant glycosylation is often correlated with inflammation and pathology such as COPD, cancer, and Crohn's disease. The inherent complexity of glycans and the diversity in the O-core structure constitute fundamental challenges for the analysis of mucin-type O-glycans. Due to coexistence of multiple isomers, multidimensional workflows such as LC-MS are required. To separate the highly polar carbohydrates, porous graphitized carbon is often used as a stationary phase. However, LC-MS workflows are time-consuming and lack reproducibility. Here we present a rapid alternative for separating and identifying O-glycans released from mucins based on trapped ion mobility mass spectrometry. Compared to established LC-MS, the acquisition time is reduced from an hour to two minutes. To test the validity, the developed workflow was applied to sputum samples from cystic fibrosis patients to map O-glycosylation features associated with disease.
Collapse
Affiliation(s)
- Leïla Bechtella
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Altensteinstraße 23A, 14195, Berlin, Germany
- Fritz Haber Institute of the Max Planck Society, Faradayweg 4‑6, 14195, Berlin, Germany
| | - Jin Chunsheng
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kerstin Fentker
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Altensteinstraße 23A, 14195, Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Güney R Ertürk
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Altensteinstraße 23A, 14195, Berlin, Germany
| | - Marc Safferthal
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Altensteinstraße 23A, 14195, Berlin, Germany
- Fritz Haber Institute of the Max Planck Society, Faradayweg 4‑6, 14195, Berlin, Germany
| | - Łukasz Polewski
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Altensteinstraße 23A, 14195, Berlin, Germany
- Fritz Haber Institute of the Max Planck Society, Faradayweg 4‑6, 14195, Berlin, Germany
| | - Michael Götze
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Altensteinstraße 23A, 14195, Berlin, Germany
- Fritz Haber Institute of the Max Planck Society, Faradayweg 4‑6, 14195, Berlin, Germany
| | - Simon Y Graeber
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Lung Research (DZL), associated partner site, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gaël M Vos
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Altensteinstraße 23A, 14195, Berlin, Germany
- Fritz Haber Institute of the Max Planck Society, Faradayweg 4‑6, 14195, Berlin, Germany
| | - Weston B Struwe
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine and Cystic Fibrosis Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Center for Lung Research (DZL), associated partner site, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Mertins
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125, Berlin, Germany
- Berlin Institute of Health, 10178, Berlin, Germany
| | - Niclas G Karlsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
| | - Kevin Pagel
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Altensteinstraße 23A, 14195, Berlin, Germany.
- Fritz Haber Institute of the Max Planck Society, Faradayweg 4‑6, 14195, Berlin, Germany.
| |
Collapse
|
5
|
Gonçalves JD, Ferraz JBS, Meirelles FV, Nociti RP, Oliveira MEF. An Exploratory Data Analysis from Ovine and Bovine RNA-Seq Identifies Pathways and Key Genes Related to Cervical Dilatation. Animals (Basel) 2023; 13:2052. [PMID: 37443850 DOI: 10.3390/ani13132052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/18/2023] [Accepted: 04/24/2023] [Indexed: 07/15/2023] Open
Abstract
The present study developed a review and exploration of data in public and already validated repositories. The main objective was to identify the pathways involved in ruminants' cervical dilatation, which are conserved between cattle and sheep in the follicular and luteal phases of the reproductive cycle. In cattle, 1961 genes were more differentially expressed in the follicular phase and 1560 in the luteal phase. An amount of 24 genes were considered exclusively expressed from these. A total of 18 genes were in the follicular phase and 6 genes were in the luteal phase. In sheep, 2126 genes were more differentially expressed in the follicular phase and 2469 genes were more differentially expressed in the luteal phase. Hoxb genes were identified in both species and are correlated with the PI3K/Akt pathway. PI3K/Akt was also found in both cattle and sheep, appearing prominently in the follicular and luteal phases of both species. Our analyses have pointed out that the PI3K/Akt pathway and the Hoxb genes appear in prominence in modulating mechanisms that involve estrus alterations in the cervix. PI3K/Akt appears to be an important pathway in the cervical relaxation process.
Collapse
Affiliation(s)
- Joedson Dantas Gonçalves
- Department of Pathology, Reproduction and One Health, School of Agricultural and Veterinarian Sciences, São Paulo State University, Via de Acesso Prof. Paulo Donato Castellane, s/n, Jaboticabal 14884-900, SP, Brazil
| | - José Bento Sterman Ferraz
- Molecular Morphophysiology and Development Laboratory, Departament of Veterinary Medicine, Faculty of Food Engineering-FZEA, University of São Paulo, Av. Duque de Caxias Norte 255, Pirassununga 14635-900, SP, Brazil
| | - Flávio Vieira Meirelles
- Molecular Morphophysiology and Development Laboratory, Departament of Veterinary Medicine, Faculty of Food Engineering-FZEA, University of São Paulo, Av. Duque de Caxias Norte 255, Pirassununga 14635-900, SP, Brazil
| | - Ricardo Perecin Nociti
- Molecular Morphophysiology and Development Laboratory, Departament of Veterinary Medicine, Faculty of Food Engineering-FZEA, University of São Paulo, Av. Duque de Caxias Norte 255, Pirassununga 14635-900, SP, Brazil
| | - Maria Emilia Franco Oliveira
- Department of Pathology, Reproduction and One Health, School of Agricultural and Veterinarian Sciences, São Paulo State University, Via de Acesso Prof. Paulo Donato Castellane, s/n, Jaboticabal 14884-900, SP, Brazil
| |
Collapse
|
6
|
Minegishi K, Dobashi Y, Koyama T, Ishibashi Y, Furuya M, Tsubochi H, Ohmoto Y, Yasuda T, Nomura S. Diagnostic utility of trefoil factor families for the early detection of lung cancer and their correlation with tissue expression. Oncol Lett 2023; 25:139. [PMID: 36909373 PMCID: PMC9996639 DOI: 10.3892/ol.2023.13725] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 12/22/2022] [Indexed: 02/23/2023] Open
Abstract
Trefoil factors (TFFs) are upregulated in numerous types of cancer, including those of the breast, the colon, the lung and the pancreas, suggesting their potential utility as biomarkers for screening. In the present study, the clinical relevance of serum or urinary TFFs as biomarkers were comprehensively evaluated and the correlation with TFF expression levels in lung cancer tissue was examined. Serum and urine were collected from 199 patients with lung cancer and 198 healthy individuals. Concentrations of serum and urinary TFF1, TFF2 and TFF3 were measured using ELISA and the potential of TFF levels to discriminate between cancer and non-cancer samples was evaluated. In 100 of the cancer cases, expression of TFF1-3 was analyzed using immunohistochemical staining of paraffin sections. Furthermore, the relationship between TFF levels and clinicopathological factors among these cancer cases was analyzed using immunohistochemistry of tissue specimens, quantified and statistically analyzed. While serum levels of all TFFs measured using ELISA were significantly higher in patients with lung cancer compared with those in healthy individuals, urinary TFFs were lower. Areas under the curve (AUC) of the receiver operating characteristic curves for serum/urinary TFF1, TFF2 and TFF3 were 0.709/0.594, 0.722/0.501 and 0.663/0.665, respectively. Furthermore, the combination of serum TFF1, TFF2, TFF3 and urinary TFF1 and TFF3 demonstrated the highest AUC (0.826). In the clinicopathological analysis, serum TFF1 was higher in the early pathological T-stage (pTis/1/2) compared with the later stage (pT3/4) and TFF2 was higher in the pN0/1 than the pN2 group. With regards to the histological types, urinary TFF1 was higher in squamous cell carcinoma than adenocarcinoma (AC), but TFF2 tended to be higher in AC. Using immunohistochemical analysis, although TFF1 and TFF3 expression showed positive correlation with serum concentrations, TFF2 was inversely correlated. In conclusion, serum and urinary TFF levels are promising predictive biomarkers, and their measurements provide a useful in vivo and non-invasive diagnostic screening tool. In particular, TFF1 and TFF3 could be surrogate markers of clinicopathological profiles of human lung cancer.
Collapse
Affiliation(s)
- Kentaro Minegishi
- Department of Thoracic Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Saitama 330-8500, Japan
| | - Yoh Dobashi
- Department of Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Saitama 330-8500, Japan.,Department of Pathology, School of Medicine, International University of Health and Welfare Hospital, Nasushiobara, Tochigi 329-2763, Japan
| | - Teruhide Koyama
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Kyoto 602-8566, Japan
| | - Yuko Ishibashi
- Department of Surgery, Breast Surgery, Tokyo Women's Medical University, Adachi Medical Center, Adachi, Tokyo 123-8558, Japan
| | - Miki Furuya
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hiroyoshi Tsubochi
- Department of Thoracic Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Saitama 330-8500, Japan
| | - Yasukazu Ohmoto
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Tokushima 770-8505, Japan
| | - Tomohiko Yasuda
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.,Department of Gastrointestinal Surgery, Nippon Medical School Chiba Hokusoh Hospital, Inzai, Chiba 270-1694, Japan
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
7
|
Qu H, Zong Q, Wang H, Wu S, Cai D, Bao W. C/EBPα Epigenetically Modulates TFF1 Expression via mC-6 Methylation in the Jejunum Inflammation Induced by a Porcine Coronavirus. Front Immunol 2022; 13:881289. [PMID: 35693767 PMCID: PMC9174463 DOI: 10.3389/fimmu.2022.881289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/26/2022] [Indexed: 11/20/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is an emerging coronavirus which causes acute diarrhea and destroys gastrointestinal barrier function in neonatal pigs. Trefoil factor 1 (TFF1) is a protective peptide for maintaining the integrity of gastrointestinal mucosa and reducing intestinal inflammation. However, its role in protecting intestinal epithelium against PEDV infection is still unclear. In this study, we discovered that TFF1 expression was activated in the jejunum of pigs with PEDV infection and TFF1 is required for the growth of porcine intestinal epithelial cells. For instance, inhibited cell proliferation and cell arrest were observed when TFF1 is genetically knocked-out using CRISPR-Cas9. Additionally, TFF1 depletion increased viral copy number and PEDV titer, along with the elevated genes involved in antiviral and inflammatory cytokines. The decreased TFF1 mRNA expression is in line with hypermethylation on the gene promoter. Notably, the strong interactions of protein-DNA complexes containing CCAAT motif significantly increased C/EBPα accessibility, whereas hypermethylation of mC-6 loci decreased C/EBPα binding occupancies in TFF1 promoter. Overall, our findings show that PEDV triggers the C/EBPα-mediated epigenetic regulation of TFF1 in intestine epithelium and facilitates host resistance to PEDV and other Coronavirus infections.
Collapse
Affiliation(s)
- Huan Qu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Qiufang Zong
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Haifei Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China
| | - Shenglong Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China
| | - Demin Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China
| |
Collapse
|
8
|
Yang H, Yang WJ, Hu B. Gastric epithelial histology and precancerous conditions. World J Gastrointest Oncol 2022; 14:396-412. [PMID: 35317321 PMCID: PMC8919001 DOI: 10.4251/wjgo.v14.i2.396] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/08/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
The most common histological type of gastric cancer (GC) is gastric adenocarcinoma arising from the gastric epithelium. Less common variants include mesenchymal, lymphoproliferative and neuroendocrine neoplasms. The Lauren scheme classifies GC into intestinal type, diffuse type and mixed type. The WHO classification includes papillary, tubular, mucinous, poorly cohesive and mixed GC. Chronic atrophic gastritis (CAG) and intestinal metaplasia are recommended as common precancerous conditions. No definite precancerous condition of diffuse/poorly/undifferentiated type is recommended. Chronic superficial inflammation and hyperplasia of foveolar cells may be the focus. Presently, the management of early GC and precancerous conditions mainly relies on endoscopy including diagnosis, treatment and surveillance. Management of precancerous conditions promotes the early detection and treatment of early GC, and even prevent the occurrence of GC. In the review, precancerous conditions including CAG, metaplasia, foveolar hyperplasia and gastric hyperplastic polyps derived from the gastric epithelium have been concluded, based on the overview of gastric epithelial histological organization and its renewal.
Collapse
Affiliation(s)
- Hang Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Wen-Juan Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bing Hu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
9
|
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
|
10
|
Sugiyama M, Machida N, Yasunaga A, Terai N, Fukasawa H, Ono HK, Kobayashi R, Nishiyama K, Hashimoto O, Kurusu S, Yoshioka K. Vaginal mucus in mice: developmental and gene expression features of epithelial mucous cells during pregnancy†. Biol Reprod 2021; 105:1272-1282. [PMID: 34416757 DOI: 10.1093/biolre/ioab157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 07/12/2021] [Accepted: 08/16/2021] [Indexed: 01/18/2023] Open
Abstract
The vagina is the site of copulation and serves as the birth canal. It also provides protection against external pathogens. In mice, due to the absence of cervical glands, the vaginal epithelium is the main producer of vaginal mucus. The development and differentiation of vaginal epithelium-constituting cells and the molecular characteristics of vaginal mucus have not been thoroughly examined. Here, we characterized vaginal mucous cell development and the expression of mucus-related factors in pregnant mice. The vaginal mucous epithelium layer thickened and became multilayered after Day 12 of pregnancy and secreted increasing amounts of mucus until early postpartum. Using histochemistry and transmission electron microscopy, we found supra-basal mucous cells as probable candidates for precursor cells. In vaginal mucous cells, the expression of TFF1, a stabilizer of mucus, was high, and some members of mucins and antimicrobial peptides (MUC5B and DEFB1) were expressed in a stage-dependent manner. In summary, this study presents the partial characterization of vaginal epithelial mucous cell lineage and expression of genes encoding several peptide substances that may affect vaginal tissue homeostasis and mucosal immunity during pregnancy and parturition.
Collapse
Affiliation(s)
- Makoto Sugiyama
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Nao Machida
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Arata Yasunaga
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan.,Department of Animal Science, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Nanako Terai
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Hanae Fukasawa
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Hisaya K Ono
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Ryosuke Kobayashi
- Laboratory of Genome Science, Biological Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Keita Nishiyama
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Osamu Hashimoto
- Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Japan
| | - Shiro Kurusu
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Kazuki Yoshioka
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan
| |
Collapse
|
11
|
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
|
12
|
Henderickx JGE, Zwittink RD, Renes IB, van Lingen RA, van Zoeren-Grobben D, Jebbink LJG, Boeren S, van Elburg RM, Knol J, Belzer C. Maturation of the preterm gastrointestinal tract can be defined by host and microbial markers for digestion and barrier defense. Sci Rep 2021; 11:12808. [PMID: 34140588 PMCID: PMC8211855 DOI: 10.1038/s41598-021-92222-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 06/01/2021] [Indexed: 12/19/2022] Open
Abstract
Functionality of the gastrointestinal tract is essential for growth and development of newborns. Preterm infants have an immature gastrointestinal tract, which is a major challenge in neonatal care. This study aims to improve the understanding of gastrointestinal functionality and maturation during the early life of preterm infants by means of gastrointestinal enzyme activity assays and metaproteomics. In this single-center, observational study, preterm infants born between 24 and 33 weeks (n = 40) and term infants born between 37 and 42 weeks (n = 3), who were admitted to Isala (Zwolle, the Netherlands), were studied. Enzyme activity analyses identified active proteases in gastric aspirates of preterm infants. Metaproteomics revealed human milk, digestive and immunological proteins in gastric aspirates of preterm infants and feces of preterm and term infants. The fecal proteome of preterm infants was deprived of gastrointestinal barrier-related proteins during the first six postnatal weeks compared to term infants. In preterm infants, bacterial oxidative stress proteins were increased compared to term infants and higher birth weight correlated to higher relative abundance of bifidobacterial proteins in postnatal week 3 to 6. Our findings indicate that gastrointestinal and beneficial microbial proteins involved in gastrointestinal maturity are associated with gestational and postnatal age.
Collapse
Affiliation(s)
- Jannie G E Henderickx
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Romy D Zwittink
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ingrid B Renes
- Danone Nutricia Research, Utrecht, the Netherlands
- Emma Children's Hospital, Amsterdam UMC, Location AMC Amsterdam, Amsterdam, The Netherlands
| | - Richard A van Lingen
- Department of Neonatology, Isala Women and Children's Hospital, Zwolle, The Netherlands
| | | | | | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University and Research, Wageningen, The Netherlands
| | - Ruurd M van Elburg
- Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan Knol
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
- Danone Nutricia Research, Utrecht, the Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
| |
Collapse
|
13
|
Yu L, Li R, Liu W, Zhou Y, Li Y, Qin Y, Chen Y, Xu Y. Protective Effects of Wheat Peptides against Ethanol-Induced Gastric Mucosal Lesions in Rats: Vasodilation and Anti-Inflammation. Nutrients 2020; 12:nu12082355. [PMID: 32784583 PMCID: PMC7469019 DOI: 10.3390/nu12082355] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
Alcohol consumption increases the risk of gastritis and gastric ulcer. Nutritional alternatives are considered for relieving the progression of gastric mucosal lesions instead of conventional drugs that produce side effects. This study was designed to evaluate the gastroprotective effects and investigate the defensive mechanisms of wheat peptides against ethanol-induced acute gastric mucosal injury in rats. Sixty male Sprague-Dawley rats were divided into six groups and orally treated with wheat peptides (0.1, 0.2, 0.4 g/kgbw) and omeprazole (20 mg/kgbw) for 4 weeks, following absolute ethanol administration for 1 h. Pretreatment with wheat peptides obviously enhanced the vasodilation of gastric mucosal blood vessels via improving the gastric mucosal blood flow and elevating the defensive factors nitric oxide (NO) and prostaglandin E2 (PGE2), and lowering the level of vasoconstrictor factor endothelin (ET)-1. Wheat peptides exhibited anti-inflammatory reaction through decreasing inducible nitric oxide synthase (iNOS) and pro-inflammatory cytokines tumor necrosis factor α (TNF-α), interleukin (IL)-1β, IL-6, and increasing trefoil factor 1 (TFF1) levels. Moreover, wheat peptides significantly down-regulated the expression of phosphorylated nuclear factor kappa-B (p-NF-κB) p65 proteins in the NF-κB signaling pathway. Altogether, wheat peptides protect gastric mucosa from ethanol-induced lesions in rats via improving the gastric microcirculation and inhibiting inflammation mediated by the NF-κB signaling transduction pathway.
Collapse
Affiliation(s)
- Lanlan Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China; (L.Y.); (R.L.); (W.L.); (Y.Z.); (Y.L.); (Y.Q.); (Y.C.)
| | - Ruijun Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China; (L.Y.); (R.L.); (W.L.); (Y.Z.); (Y.L.); (Y.Q.); (Y.C.)
| | - Wei Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China; (L.Y.); (R.L.); (W.L.); (Y.Z.); (Y.L.); (Y.Q.); (Y.C.)
| | - Yalin Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China; (L.Y.); (R.L.); (W.L.); (Y.Z.); (Y.L.); (Y.Q.); (Y.C.)
| | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China; (L.Y.); (R.L.); (W.L.); (Y.Z.); (Y.L.); (Y.Q.); (Y.C.)
| | - Yong Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China; (L.Y.); (R.L.); (W.L.); (Y.Z.); (Y.L.); (Y.Q.); (Y.C.)
| | - Yuhan Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China; (L.Y.); (R.L.); (W.L.); (Y.Z.); (Y.L.); (Y.Q.); (Y.C.)
| | - Yajun Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100083, China; (L.Y.); (R.L.); (W.L.); (Y.Z.); (Y.L.); (Y.Q.); (Y.C.)
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100083, China
- Correspondence: ; Tel.: +86-10-8280-2552
| |
Collapse
|
14
|
Heuer J, Heuer F, Stürmer R, Harder S, Schlüter H, Braga Emidio N, Muttenthaler M, Jechorek D, Meyer F, Hoffmann W. The Tumor Suppressor TFF1 Occurs in Different Forms and Interacts with Multiple Partners in the Human Gastric Mucus Barrier: Indications for Diverse Protective Functions. Int J Mol Sci 2020; 21:ijms21072508. [PMID: 32260357 PMCID: PMC7177788 DOI: 10.3390/ijms21072508] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/30/2020] [Accepted: 04/02/2020] [Indexed: 12/15/2022] Open
Abstract
TFF1 is a protective peptide of the Trefoil Factor Family (TFF), which is co-secreted with the mucin MUC5AC, gastrokine 2 (GKN2), and IgG Fc binding protein (FCGBP) from gastric surface mucous cells. Tff1-deficient mice obligatorily develop antropyloric adenoma and about 30% progress to carcinomas, indicating that Tff1 is a tumor suppressor. As a hallmark, TFF1 contains seven cysteine residues with three disulfide bonds stabilizing the conserved TFF domain. Here, we systematically investigated the molecular forms of TFF1 in the human gastric mucosa. TFF1 mainly occurs in an unusual monomeric form, but also as a homodimer. Furthermore, minor amounts of TFF1 form heterodimers with GKN2, FCGBP, and an unknown partner protein, respectively. TFF1 also binds to the mucin MUC6 in vitro, as shown by overlay assays with synthetic 125I-labeled TFF1 homodimer. The dominant presence of a monomeric form with a free thiol group at Cys-58 is in agreement with previous studies in Xenopus laevis and mouse. Cys-58 is likely highly reactive due to flanking acid residues (PPEEEC58EF) and might act as a scavenger for extracellular reactive oxygen/nitrogen species protecting the gastric mucosa from damage by oxidative stress, e.g., H2O2 generated by dual oxidase (DUOX).
Collapse
Affiliation(s)
- Jörn Heuer
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Franziska Heuer
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - René Stürmer
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Sönke Harder
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Hartmut Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Nayara Braga Emidio
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Markus Muttenthaler
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Dörthe Jechorek
- Institute of Pathology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Frank Meyer
- Department of Surgery, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
- Correspondence:
| |
Collapse
|
15
|
Liu F, Fu J, Bergstrom K, Shan X, McDaniel JM, McGee S, Bai X, Chen W, Xia L. Core 1-derived mucin-type O-glycosylation protects against spontaneous gastritis and gastric cancer. J Exp Med 2020; 217:e20182325. [PMID: 31645367 PMCID: PMC7037257 DOI: 10.1084/jem.20182325] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 08/12/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022] Open
Abstract
Core 1-derived mucin-type O-glycans (O-glycans) are a major component of gastric mucus with an unclear role. To address this, we generated mice lacking gastric epithelial O-glycans (GEC C1galt1-/-). GEC C1galt1-/- mice exhibited spontaneous gastritis that progressed to adenocarcinoma with ∼80% penetrance by 1 yr. GEC C1galt1-/- gastric epithelium exhibited defective expression of a major mucus forming O-glycoprotein Muc5AC relative to WT controls, which was associated with impaired gastric acid homeostasis. Inflammation and tumorigenesis in GEC C1galt1-/- stomach were concurrent with activation of caspases 1 and 11 (Casp1/11)-dependent inflammasome. GEC C1galt1-/- mice genetically lacking Casp1/11 had reduced gastritis and gastric cancer progression. Notably, expression of Tn antigen, a truncated form of O-glycan, and CASP1 activation was associated with tumor progression in gastric cancer patients. These results reveal a critical role of O-glycosylation in gastric homeostasis and the protection of the gastric mucosa from Casp1-mediated gastric inflammation and cancer.
Collapse
Affiliation(s)
- Fei Liu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Jianxin Fu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Kirk Bergstrom
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Xindi Shan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - J. Michael McDaniel
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Samuel McGee
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Xia Bai
- Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
16
|
Popp J, Schicht M, Garreis F, Klinger P, Gelse K, Sesselmann S, Tsokos M, Etzold S, Stiller D, Claassen H, Paulsen F. Human Synovia Contains Trefoil Factor Family (TFF) Peptides 1-3 Although Synovial Membrane Only Produces TFF3: Implications in Osteoarthritis and Rheumatoid Arthritis. Int J Mol Sci 2019; 20:ijms20236105. [PMID: 31817054 PMCID: PMC6928748 DOI: 10.3390/ijms20236105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 01/15/2023] Open
Abstract
Objective: Trefoil factor family peptide 3 (TFF3) has been shown to support catabolic functions in cases of osteoarthritis (OA). As in joint physiology and diseases such as OA, the synovial membrane (SM) of the joint capsule also plays a central role. We analyze 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.
Collapse
Affiliation(s)
- Judith Popp
- Friedrich Alexander University Erlangen-Nürnberg (FAU), Institute of Functional and Clinical Anatomy, 91054 Erlangen, Germany; (J.P.); (M.S.); (F.G.); (P.K.)
| | - Martin Schicht
- Friedrich Alexander University Erlangen-Nürnberg (FAU), Institute of Functional and Clinical Anatomy, 91054 Erlangen, Germany; (J.P.); (M.S.); (F.G.); (P.K.)
| | - Fabian Garreis
- Friedrich Alexander University Erlangen-Nürnberg (FAU), Institute of Functional and Clinical Anatomy, 91054 Erlangen, Germany; (J.P.); (M.S.); (F.G.); (P.K.)
| | - Patricia Klinger
- Friedrich Alexander University Erlangen-Nürnberg (FAU), Institute of Functional and Clinical Anatomy, 91054 Erlangen, Germany; (J.P.); (M.S.); (F.G.); (P.K.)
| | - Kolja Gelse
- University Hospital Erlangen, Department of Trauma Surgery, 91054 Erlangen, Germany;
| | - Stefan Sesselmann
- University of Applied Sciences Amberg-Weiden, Institute for Medical Engineering, 92637 Weiden, Germany;
| | - Michael Tsokos
- Charité-Universitätsmedizin Berlin, Institute of Legal Medicine and Forensic Sciences, 10117 Berlin, Germany; (M.T.); (S.E.)
| | - Saskia Etzold
- Charité-Universitätsmedizin Berlin, Institute of Legal Medicine and Forensic Sciences, 10117 Berlin, Germany; (M.T.); (S.E.)
| | - Dankwart Stiller
- Martin Luther University Halle-Wittenberg (MLU), Department of Legal Medicine, 06108 Halle (Saale), Germany;
| | - Horst Claassen
- Martin Luther University Halle-Wittenberg (MLU), Department of Anatomy and Cell Biology, 06108 Halle (Saale), Germany;
| | - Friedrich Paulsen
- Friedrich Alexander University Erlangen-Nürnberg (FAU), Institute of Functional and Clinical Anatomy, 91054 Erlangen, Germany; (J.P.); (M.S.); (F.G.); (P.K.)
- Sechenov University, Department of Topographic Anatomy and Operative Surgery, 119146 Moscow, Russia
- Correspondence: ; Tel.: +49-9131-8522865; Fax: +49-9131-8522862
| |
Collapse
|
17
|
Muc5ac null mice are predisposed to spontaneous gastric antro-pyloric hyperplasia and adenomas coupled with attenuated H. pylori-induced corpus mucous metaplasia. J Transl Med 2019; 99:1887-1905. [PMID: 31399638 PMCID: PMC6927550 DOI: 10.1038/s41374-019-0293-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 12/15/2022] Open
Abstract
Gastric cancer (GC) is the third leading cause of cancer-related deaths worldwide and is strongly associated with chronic Helicobacter pylori (Hp) infection. The ability of Hp to closely adhere to the gastric surface protective mucous layer containing mucins (MUC in humans and Muc in animals), primarily Muc5ac, is integral in the stepwise pathogenesis from gastritis to cancer. To probe the role of Muc5ac in Hp-induced gastric pathology, Muc5ac-/- and Muc5ac+/+ (WT) mice were experimentally infected with Hp Sydney strain (SS1). At 16 weeks and 32 weeks post infection (wpi), groups of mice were euthanized and evaluated for the following: gastric histopathological parameters, immunohistochemical expression of mucins (Muc5ac, Muc1, Muc2), Trefoil factor family proteins (Tff1 and Tff2), Griffonia (Bandeiraea) simplicifolia lectin II (GSL II) (mucous metaplasia marker) and Clusterin (Spasmolytic Polypeptide Expressing Metaplasia (SPEM) marker), Hp colonization density by qPCR and gastric cytokine mRNA levels. Our results demonstrate that Muc5ac-/- mice developed spontaneous antro-pyloric proliferation, adenomas and in one case with neuroendocrine differentiation; these findings were independent of Hp infection along with strong expression levels of Tff1, Tff2 and Muc1. Hp-infected Muc5ac-/- mice had significantly lowered gastric corpus mucous metaplasia at 16 wpi and 32 wpi (P = 0.0057 and P = 0.0016, respectively), with a slight reduction in overall gastric corpus pathology. GSII-positive mucous neck cells were decreased in Hp-infected Muc5ac-/- mice compared to WT mice and clusterin positivity was noted within metaplastic glands in both genotypes following Hp infection. Additionally, Hp colonization densities were significantly higher in Muc5ac-/- mice compared to WT at 16 wpi in both sexes (P = 0.05) along with a significant reduction in gastric Tnfα (16 wpi-males and females, P = 0.017 and P = 0.036, respectively and 32 wpi-males only, P = 0.025) and Il-17a (16 wpi-males) (P = 0.025). Taken together, our findings suggest a protective role for MUC5AC/Muc5ac in maintaining gastric antral equilibrium and inhibiting Hp colonization and associated inflammatory pathology.
Collapse
|
18
|
The Interaction of Helicobacter pylori with TFF1 and Its Role in Mediating the Tropism of the Bacteria Within the Stomach. Int J Mol Sci 2019; 20:ijms20184400. [PMID: 31500233 PMCID: PMC6769565 DOI: 10.3390/ijms20184400] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/27/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori colonises the human stomach and has tropism for the gastric mucin, MUC5AC. The majority of organisms live in the adherent mucus layer within their preferred location, close to the epithelial surface where the pH is near neutral. Trefoil factor 1 (TFF1) is a small trefoil protein co-expressed with the gastric mucin MUC5AC in surface foveolar cells and co-secreted with MUC5AC into gastric mucus. Helicobacter pylori binds with greater avidity to TFF1 dimer, which is present in gastric mucus, than to TFF1 monomer. Binding of H. pylori to TFF1 is mediated by the core oligosaccharide subunit of H. pylori lipopolysaccharide at pH 5.0–6.0. Treatment of H. pylori lipopolysaccharide with mannosidase or glucosidase inhibits its interaction with TFF1. Both TFF1 and H. pylori have a propensity for binding to mucins with terminal non-reducing α- or β-linked N-acetyl-d-glucosamine or α-(2,3) linked sialic acid or Gal-3-SO42−. These findings are strong evidence that TFF1 has carbohydrate-binding properties that may involve a conserved patch of aromatic hydrophobic residues on the surface of its trefoil domain. The pH-dependent lectin properties of TFF1 may serve to locate H. pylori deep in the gastric mucus layer close to the epithelium rather than at the epithelial surface. This restricted localisation could limit the interaction of H. pylori with epithelial cells and the subsequent host signalling events that promote inflammation.
Collapse
|
19
|
Soutto M, Chen Z, Bhat AA, Wang L, Zhu S, Gomaa A, Bates A, Bhat NS, Peng D, Belkhiri A, Piazuelo MB, Washington MK, Steven XC, Peek R, El-Rifai W. Activation of STAT3 signaling is mediated by TFF1 silencing in gastric neoplasia. Nat Commun 2019; 10:3039. [PMID: 31292446 PMCID: PMC6620282 DOI: 10.1038/s41467-019-11011-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 06/12/2019] [Indexed: 01/01/2023] Open
Abstract
TFF1, a secreted protein, plays an essential role in keeping the integrity of gastric mucosa and its barrier function. Loss of TFF1 expression in the TFF1-knockout (KO) mouse leads to a pro-inflammatory phenotype with a cascade of gastric lesions that include low-grade dysplasia, high-grade dysplasia, and adenocarcinomas. In this study, we demonstrate nuclear localization of p-STATY705, with significant overexpression of several STAT3 target genes in gastric glands from the TFF1-KO mice. We also show frequent loss of TFF1 with nuclear localization of STAT3 in human gastric cancers. The reconstitution of TFF1 protein in human gastric cancer cells and 3D gastric glands organoids from TFF1-KO mice abrogates IL6-induced nuclear p-STAT3Y705 expression. Reconstitution of TFF1 inhibits IL6-induced STAT3 transcription activity, suppressing expression of its target genes. TFF1 blocks IL6Rα-GP130 complex formation through interfering with binding of IL6 to its receptor IL6Rα. These findings demonstrate a functional role of TFF1 in suppressing gastric tumorigenesis by impeding the IL6-STAT3 pro-inflammatory signaling axis. Trefoil factor 1 (TFF1) is a protein secreted by the gastric mucosa that protects against gastric tumourigenesis. Here, the authors show that TFF1 inhibits the oncogenic inflammatory response and IL-6-mediated STAT3 activation by interfering with the binding of IL6 to its receptor IL6Rα.
Collapse
Affiliation(s)
- Mohammed Soutto
- Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA.,Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Zheng Chen
- Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA.,Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ajaz A Bhat
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.,Division of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Lihong Wang
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shoumin Zhu
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ahmed Gomaa
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andreia Bates
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nadeem S Bhat
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dunfa Peng
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Abbes Belkhiri
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Blanca Piazuelo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Kay Washington
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xi Chen Steven
- Department of Public Health Sciences, Division of Biostatistics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Richard Peek
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wael El-Rifai
- Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA. .,Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA. .,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
20
|
Binding of Helicobacter pylori to Human Gastric Mucins Correlates with Binding of TFF1. Microorganisms 2018; 6:microorganisms6020044. [PMID: 29783620 PMCID: PMC6027488 DOI: 10.3390/microorganisms6020044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/23/2018] [Accepted: 05/01/2018] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori binds to the gastric mucin, MUC5AC, and to trefoil factor, TFF1, which has been shown to interact with gastric mucin. We examined the interactions of TFF1 and H. pylori with purified gastrointestinal mucins from different animal species and from humans printed on a microarray platform to investigate whether TFF1 may play a role in locating H. pylori in gastric mucus. TFF1 bound almost exclusively to human gastric mucins and did not interact with human colonic mucins. There was a strong correlation between binding of TFF1 and H. pylori to human gastric mucins, and between binding of both TFF1 and H. pylori to gastric mucins with that of Griffonia simplicifolia lectin-II, which is specific for terminal non-reducing α- or β-linked N-acetyl-d-glucosamine. These results suggest that TFF1 may help to locate H. pylori in a discrete layer of gastric mucus and hence restrain their interactions with epithelial cells.
Collapse
|
21
|
Tang L, Chen Q, Sun L, Zhu L, Liu J, Meng Z, Ni Z, Wang X. Curcumin suppresses MUC5AC production via interfering with the EGFR signaling pathway. Int J Mol Med 2018; 42:497-504. [PMID: 29620257 DOI: 10.3892/ijmm.2018.3609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 03/23/2018] [Indexed: 11/06/2022] Open
Abstract
Excessive mucin production in the airway may contribute to airway inflammatory diseases. Curcumin has been reported to prevent mucin 5AC (MUC5AC) production in human airway epithelial cells; however, the molecular targets of curcumin involved in regulating MUC5AC expression have remained elusive. The present study aimed to elucidate the molecular mechanisms by which curcumin regulates MUC5AC production, utilizing the NCI‑H292 human airway epithelial cell line featuring MUC5AC hypersecretion. Curcumin was able to counteract the endothelial growth factor (EGF)‑stimulated mRNA and protein expression of MUC5AC. In addition, curcumin treatment prevented EGF‑induced phosphorylation of EGF receptor (EGFR) as well as the downstream AKT and signal transducer and activator of transcription 3 (STAT3), while inhibition of PI3K and STAT3 signaling significantly attenuated the expression of MUC5AC that was induced by EGF. Furthermore, EGF‑induced increases in the levels of phosphorylated STAT3 in the nuclear fraction were inhibited by curcumin and PI3K inhibitors. In addition, treatment with curcumin significantly decreased MUC5AC and EGFR expression in a time‑dependent manner under basal conditions. These results demonstrated that curcumin inhibited MUC5AC protein expression in NCI‑H292 cells under basal conditions as well under EGF stimulation. This inhibition was accompanied by decreased activation of the EGFR/AKT/STAT3 pathway and reduced EGFR expression, which indicated that curcumin may have a dual role in interfering with the EGFR signaling pathway and inhibiting mucin expression in human airway epithelial cells.
Collapse
Affiliation(s)
- Lingling Tang
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Qingge Chen
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Li Sun
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Linyun Zhu
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Jinjin Liu
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Ziyu Meng
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Zhenhua Ni
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Xiongbiao Wang
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| |
Collapse
|
22
|
Demouveaux B, Gouyer V, Gottrand F, Narita T, Desseyn JL. Gel-forming mucin interactome drives mucus viscoelasticity. Adv Colloid Interface Sci 2018; 252:69-82. [PMID: 29329667 DOI: 10.1016/j.cis.2017.12.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 12/31/2022]
Abstract
Mucus is a hydrogel that constitutes the first innate defense in all mammals. The main organic component of mucus, gel-forming mucins, forms a complex network through both reversible and irreversible interactions that drive mucus gel formation. Significant advances in the understanding of irreversible gel-forming mucins assembly have been made using recombinant protein approaches. However, little is known about the reversible interactions that may finely modulate mucus viscoelasticity, which can be characterized using rheology. This approach can be used to investigate both the nature of gel-forming mucins interactions and factors that influence hydrogel formation. This knowledge is directly relevant to the development of new drugs to modulate mucus viscoelasticity and to restore normal mucus functions in diseases such as in cystic fibrosis. The aim of the present review is to summarize the current knowledge about the relationship between the mucus protein matrix and its functions, with emphasis on mucus viscoelasticity.
Collapse
Affiliation(s)
| | - Valérie Gouyer
- Univ. Lille, Inserm, CHU Lille, LIRIC UMR 995, F-59000 Lille, France
| | - Frédéric Gottrand
- Univ. Lille, Inserm, CHU Lille, LIRIC UMR 995, F-59000 Lille, France
| | - Tetsuharu Narita
- Laboratoire Sciences et Ingénierie de la Matière Molle, PSL Research University, UPMC Univ Paris 06, ESPCI Paris, CNRS, 10 rue Vauquelin, 75231 Paris Cedex 05, France; Global Station for Soft Matter, Global Institution for Collaborative Research and Education, Hokkaido University, Sapporo, Japan
| | - Jean-Luc Desseyn
- Univ. Lille, Inserm, CHU Lille, LIRIC UMR 995, F-59000 Lille, France.
| |
Collapse
|
23
|
Abstract
Trefoil factor (TFF) peptides, with a 40-amino acid motif and including six conserved cysteine residues that form intramolecular disulfide bonds, are a family of mucin-associated secretory molecules mediating many physiological roles that maintain and restore gastrointestinal (GI) mucosal homeostasis. TFF peptides play important roles in response to GI mucosal injury and inflammation. In response to acute GI mucosal injury, TFF peptides accelerate cell migration to seal the damaged area from luminal contents, whereas chronic inflammation leads to increased TFF expression to prevent further progression of disease. Although much evidence supports the physiological significance of TFF peptides in mucosal defenses, the molecular and cellular mechanisms of TFF peptides in the GI epithelium remain largely unknown. In this review, we summarize the functional roles of TFF1, 2, and 3 and illustrate their action mechanisms, focusing on defense mechanisms in the GI tract.
Collapse
Affiliation(s)
- Eitaro Aihara
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267;
| | - Kristen A Engevik
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267;
| | - Marshall H Montrose
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267;
| |
Collapse
|
24
|
Bastholm SK, Samson MH, Becher N, Hansen LK, Stubbe PR, Chronakis IS, Nexo E, Uldbjerg N. Trefoil factor peptide 3 is positively correlated with the viscoelastic properties of the cervical mucus plug. Acta Obstet Gynecol Scand 2016; 96:47-52. [PMID: 27731893 DOI: 10.1111/aogs.13038] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 10/07/2016] [Indexed: 11/27/2022]
Abstract
INTRODUCTION The viscoelastic properties of the cervical mucus plug are considered essential for the occlusion of the cervical canal and thereby for protection against ascending infections during pregnancy. Factors controlling this property are virtually unknown. This study explores a possible role of trefoil factor peptides 1, 2 and 3 (TFF1-3); peptides believed to influence mucus viscosity. MATERIAL AND METHODS The study is based on spontaneously shed cervical mucus plugs from 14 women in active labor. The viscoelastic properties; the elastic modulus (G') and the viscous modulus (G") were determined by an oscillatory rheometer. The concentrations of TFF1-3 were measured by an in-house enzyme-linked immunosorbent assay. Associations were analyzed by random-effects generalized least-squares regression analyses. RESULTS Median (range) concentrations of TFF1, TFF2 and TFF3 were 3.1 (1.2-8.6), 1.1 (<0.006-3.7) and 1000 (170-5300) nmol/g cervical mucus plug, respectively. The TFF3 concentration was associated with G' (regression coefficient 11.7 Pa/Log nm; 95% CI 3.0-20.4, p = 0.009) and G" (regression coefficient 3.2 Pa/Log nm; 95% CI 1.5-5.0, p < 0.001). CONCLUSION We suggest that TFF3 plays a role in the viscoelastic properties of the cervical mucus plug.
Collapse
Affiliation(s)
- Sara K Bastholm
- Department of Obstetrics and Gynecology, Aarhus University Hospital, Aarhus, Denmark
| | - Mie H Samson
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Naja Becher
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| | - Lea K Hansen
- Department of Obstetrics and Gynecology, Aarhus University Hospital, Aarhus, Denmark
| | - Peter R Stubbe
- Food Production Engineering Research Group, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Ioannis S Chronakis
- Nano-BioScience Research Group, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Ebba Nexo
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Niels Uldbjerg
- Department of Obstetrics and Gynecology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
25
|
May FEB, Westley BR. TFF3 is a valuable predictive biomarker of endocrine response in metastatic breast cancer. Endocr Relat Cancer 2015; 22:465-79. [PMID: 25900183 PMCID: PMC4455223 DOI: 10.1530/erc-15-0129] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2015] [Indexed: 12/13/2022]
Abstract
The stratification of breast cancer patients for endocrine therapies by oestrogen or progesterone receptor expression is effective but imperfect. The present study aims were to validate microarray studies that demonstrate TFF3 regulation by oestrogen and its association with oestrogen receptors in breast cancer, to evaluate TFF3 as a biomarker of endocrine response, and to investigate TFF3 function. Microarray data were validated by quantitative RT-PCR and northern and western transfer analyses. TFF3 was induced by oestrogen, and its induction was inhibited by antioestrogens, tamoxifen, 4-hydroxytamoxifen and fulvestrant in oestrogen-responsive breast cancer cells. The expression of TFF3 mRNA was associated with oestrogen receptor mRNA in breast tumours (Pearson's coefficient=0.762, P=0.000). Monoclonal antibodies raised against the TFF3 protein detected TFF3 by immunohistochemistry in oesophageal submucosal glands, intestinal goblet and neuroendocrine cells, Barrett's metaplasia and intestinal metaplasia. TFF3 protein expression was associated with oestrogen receptor, progesterone receptor and TFF1 expression in malignant breast cells. TFF3 is a specific and sensitive predictive biomarker of response to endocrine therapy, degree of response and duration of response in unstratified metastatic breast cancer patients (P=0.000, P=0.002 and P=0.002 respectively). Multivariate binary logistic regression analysis demonstrated that TFF3 is an independent biomarker of endocrine response and degree of response, and this was confirmed in a validation cohort. TFF3 stimulated migration and invasion of breast cancer cells. In conclusion, TFF3 expression is associated with response to endocrine therapy, and outperforms oestrogen receptor, progesterone receptor and TFF1 as an independent biomarker, possibly because it mediates the malign effects of oestrogen on invasion and metastasis.
Collapse
Affiliation(s)
- Felicity E B May
- Department of Pathology Faculty of Medical Sciences, Northern Institute for Cancer Research and Newcastle University Institute for Ageing, University of Newcastle-upon-Tyne, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Bruce R Westley
- Department of Pathology Faculty of Medical Sciences, Northern Institute for Cancer Research and Newcastle University Institute for Ageing, University of Newcastle-upon-Tyne, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| |
Collapse
|
26
|
Cheng YM, Lu MT, Yeh CM. Functional expression of recombinant human trefoil factor 1 by Escherichia coli and Brevibacillus choshinensis. BMC Biotechnol 2015; 15:32. [PMID: 25990322 PMCID: PMC4438461 DOI: 10.1186/s12896-015-0149-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 04/22/2015] [Indexed: 12/13/2022] Open
Abstract
Background Trefoil factor 1 (TFF1) mediates mucosal repair and belongs to a highly conserved trefoil factor family proteins which are secreted by epithelial cells in the stomach or colon mucous membrane. TFF1 forms a homodimer via a disulphide linkage that affects wound healing activity. Previous recombinant expressions of TFF1 were too low yield for industrial application. This study aims to improve the expression level of bioactive recombinant TFF1 (rTFF1) and facilitate application potency. Methods The rTFF1 gene rtff1 was synthesized, expressed by Escherichia coli and secreted by Brevibacillus choshinensis. The rTFF1s were purified. The polymeric patterns and wound healing capacities of purified rTFF1s were checked. Results In Escherichia coli, 21.08 mg/L rTFF1 was stably expressed as monomer, dimer and oligomer in soluble fraction. In Brevebacillus choshinensis, the rTFF1 was secreted extracellularly at high level (35.73 mg/L) and formed monomer, dimer and oligomer forms. Both proteins from different sources were purified by Ni-NTA chromatography and exhibited the wound healing activities. The rTFF1 produced by B. choshinensis had better wound healing capability than the rTFF1 produced by E. coli. After pH 2.4 buffer treatments, the purified rTFF1 formed more oligomeric forms as well as better wound healing capability. Glycosylation assay and LC-MS/MS spectrometry experiments showed that the rTFF1 produced by B. choshinensis was unexpectedly glycosylated at N-terminal Ser residue. The glycosylation may contribute to the better wound healing capacity. Conclusions This study provides a potent tool of rTFF1 production to be applied in gastric damage protection and wound healing. The protein sources from B. choshinensis were more efficient than rTFF1 produced by E. coli. Electronic supplementary material The online version of this article (doi:10.1186/s12896-015-0149-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yueh-Mei Cheng
- Department of Food Science and Biotechnology, National Chung-Hsing University, Taichung, Taiwan, Republic of China.
| | - Meng-Ting Lu
- Department of Food Science and Biotechnology, National Chung-Hsing University, Taichung, Taiwan, Republic of China.
| | - Chuan Mei Yeh
- Department of Food Science and Biotechnology, National Chung-Hsing University, Taichung, Taiwan, Republic of China. .,Agricultural Biotechnology Center, National Chung-Hsing University, Taichung, Taiwan, Republic of China.
| |
Collapse
|
27
|
Viby NE, Pedersen L, Lund TK, Kissow H, Backer V, Nexø E, Thim L, Poulsen SS. Trefoil factor peptides in serum and sputum from subjects with asthma and COPD. CLINICAL RESPIRATORY JOURNAL 2014; 9:322-9. [PMID: 24720774 DOI: 10.1111/crj.12146] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 02/23/2014] [Accepted: 04/04/2014] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Trefoil factor peptides (TFF) are secreted onto mucosal surfaces together with mucins and occur in high concentrations in pulmonary secretions from patients with chronic obstructive pulmonary disease (COPD). In the present study, we aimed to explore the concentrations of the peptides in serum and sputum in patients with COPD. MATERIALS AND METHODS Thirty-five individuals were included in the study, including 11 healthy individuals, 13 indivials with asthma and 11 individuals with COPD. TFF1, TFF2 and TFF3 were measured by enzyme-linked immunosorbent assay (ELISA) in sputum induced by hypertonic saline inhalation and in serum. Total protein content in sputum was also determined. RESULTS In the sputum samples from COPD patients, we observed an eightfold higher concentration of TFF1 and a fivefold higher concentration of TFF3 compared with controls. In the serum samples from COPD patients, we observed three-, three- and twofold higher concentrations of TFF1, TFF2 and TFF3 respectively compared with controls. CONCLUSIONS There is increased secretion of TFF peptides in the lungs of patients with COPD, as well as significant increases in serum levels. This suggests a role for TFF peptides in the pathogenesis of pulmonary diseases with mucus hypersecretion.
Collapse
Affiliation(s)
- Niels-Erik Viby
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Cardiothoracic Surgery, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lars Pedersen
- Department of Respiratory Medicine, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Thomas Kromann Lund
- Department of Respiratory Medicine, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Hannelouise Kissow
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vibeke Backer
- Department of Respiratory Medicine, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Ebba Nexø
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Lars Thim
- Department of Protein Engineering, Novo Nordisk A/S, Maalov, Denmark
| | - Steen Seier Poulsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Dai J, Zhang N, Wang J, Chen M, Chen J. Gastrokine-2 is downregulated in gastric cancer and its restoration suppresses gastric tumorigenesis and cancer metastasis. Tumour Biol 2014; 35:4199-207. [DOI: 10.1007/s13277-013-1550-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 12/13/2013] [Indexed: 12/22/2022] Open
|
29
|
Bastholm SK, Becher N, Stubbe PR, Chronakis IS, Uldbjerg N. The viscoelastic properties of the cervical mucus plug. Acta Obstet Gynecol Scand 2014; 93:201-8. [DOI: 10.1111/aogs.12308] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Sara K. Bastholm
- Department of Obstetrics and Gynecology; Aarhus University Hospital; Aarhus Denmark
| | - Naja Becher
- Department of Clinical Genetics; Aarhus University Hospital; Aarhus Denmark
| | | | | | - Niels Uldbjerg
- Department of Obstetrics and Gynecology; Aarhus University Hospital; Aarhus Denmark
| |
Collapse
|
30
|
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
|
31
|
In silico analysis of stomach lineage specific gene set expression pattern in gastric cancer. Biochem Biophys Res Commun 2013; 439:539-46. [DOI: 10.1016/j.bbrc.2013.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 09/02/2013] [Indexed: 01/28/2023]
|
32
|
Activation of the NF-kB pathway downregulates TFF-1 in gastric carcinogenesis. Virchows Arch 2013; 463:497-507. [PMID: 23942618 DOI: 10.1007/s00428-013-1469-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 07/15/2013] [Accepted: 08/02/2013] [Indexed: 02/06/2023]
Abstract
Trefoil factor 1 (TFF1) is expressed in the normal superficial epithelium of the stomach and is implicated in the maintenance of gastric epithelial structure and function. During gastric carcinogenesis, in which pro-inflammatory cytokines play a crucial role, its expression level decreases suggesting a role as tumor suppressor factor. We have compared expression of TFF1 in gastric mucosa from cancer patients, in which several degrees of inflammatory infiltrate are present, with that in normal mucosa from non-cancer patients without infiltrating inflammatory cells. TFF1 is less expressed in the superficial gastric epithelium from cancer patients than in that from normal individuals in which the nuclear factor (NF)-κB pathway is not activated. We analyzed TFF1 expression in ex vivo samples of gastric mucosa from cancer patients, and in MKN45 gastric cancer cell line after exposure to proinflammatory cytokines interleukin (IL)-1β or tumor necrosis factor (TNF)-α, that activate the NF-κB pathway. We found that IL-1β and TNF-α activate the NF-κB pathway, as reflected in the nuclear expression of p65 and the activation of p-IκBα, and downregulate TFF1 expression after 1 or 2 h of exposure. Moreover, cells in the superficial gastric epithelium in ex vivo samples co-expressed TFF1/p65 at cellular level, whereas tumor cells did not. In summary, downregulation of TFF1 expression during gastric neoplastic transformation is associated with activation of the NF-κB pathway through IL-1β or TNF-α, but other regulatory mechanisms might also be involved.
Collapse
|
33
|
Virulent Shigella flexneri affects secretion, expression, and glycosylation of gel-forming mucins in mucus-producing cells. Infect Immun 2013; 81:3632-43. [PMID: 23876800 DOI: 10.1128/iai.00551-13] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mucin glycoproteins are secreted in large amounts by the intestinal epithelium and constitute an efficient component of innate immune defenses to promote homeostasis and protect against enteric pathogens. In this study, our objective was to investigate how the bacterial enteropathogen Shigella flexneri, which causes bacillary dysentery, copes with the mucin defense barrier. We report that upon in vitro infection of mucin-producing polarized human intestinal epithelial cells, virulent S. flexneri manipulates the secretion of gel-forming mucins. This phenomenon, which is triggered only by virulent strains, results in accumulation of mucins at the cell apical surface, leading to the appearance of a gel-like structure that favors access of bacteria to the cell surface and the subsequent invasion process. We identify MUC5AC, a gel-forming mucin, as a component of this structure. Formation of this gel does not depend on modifications of electrolyte concentrations, induction of trefoil factor expression, endoplasmic reticulum stress, or response to unfolded proteins. In addition, transcriptional and biochemical analyses of infected cells reveal modulations of mucin gene expression and modifications of mucin glycosylation patterns, both of which are induced by virulent bacteria in a type III secretion system-dependent manner. Thus, S. flexneri has developed a dedicated strategy to alter the mucus barrier by targeting key elements of the gel-forming capacity of mucins: gene transcription, protein glycosylation, and secretion.
Collapse
|
34
|
PELDEN SONAM, INSAWANG TONKLA, THUWAJIT CHANITRA, THUWAJIT PETI. The trefoil factor 1 (TFF1) protein involved in doxorubicin-induced apoptosis resistance is upregulated by estrogen in breast cancer cells. Oncol Rep 2013; 30:1518-26. [DOI: 10.3892/or.2013.2593] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 06/26/2013] [Indexed: 11/05/2022] Open
|
35
|
Samson MH. Quantitative measurements of trefoil factor family peptides: possibilities and pitfalls. Scandinavian Journal of Clinical and Laboratory Investigation 2013; 73:193-202. [PMID: 23391285 DOI: 10.3109/00365513.2013.765962] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The trefoil factor family (TFF) peptides TFF1, TFF2, and TFF3 are produced and secreted by mucous membranes throughout the body. Their importance for the protection and repair of epithelial surfaces is well established, and the three peptides are present in various amounts in mucosal secretions as well as in the circulation. They have been linked to both inflammatory diseases and to various types of cancer, and serum concentrations of TFF3 show a more than 47-fold increase during pregnancy. Several both commercial and in-house immunoassays exist, but a number of methodological issues remain unresolved. This review describes methodological challenges in the measurement of the peptides in humans, and summarizes current knowledge concerning the occurrence and possible significance of the peptides in human health and disease.
Collapse
|
36
|
Pluta K, McGettigan PA, Reid CJ, Browne JA, Irwin JA, Tharmalingam T, Corfield A, Baird A, Loftus BJ, Evans ACO, Carrington SD. Molecular aspects of mucin biosynthesis and mucus formation in the bovine cervix during the periestrous period. Physiol Genomics 2012; 44:1165-78. [DOI: 10.1152/physiolgenomics.00088.2012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mucus within the cervical canal represents a hormonally regulated barrier that reconciles the need to exclude the vaginal microflora from the uterus during progesterone dominance, while permitting sperm transport at estrus. Its characteristics change during the estrous cycle to facilitate these competing functional requirements. Hydrated mucin glycoproteins synthesized by the endocervical epithelium form the molecular scaffold of this mucus. This study uses the bovine cervix as a model to examine functional groups of genes related to mucin biosynthesis and mucus production over the periestrous period when functional changes in cervical barrier function are most prominent. Cervical tissue samples were collected from 30 estrus synchronized beef heifers. Animals were slaughtered in groups starting 12 h after the withdrawal of intravaginal progesterone releasing devices (controlled internal drug releases) until 7 days postonset of estrus (luteal phase). Subsequent groupings represented proestrus, early estrus, late estrus, metestrus, and finally the early luteal phase. Tissues were submitted to next generation RNA-seq transcriptome analysis. We identified 114 genes associated with biosynthesis and intracellular transport of mucins, and postsecretory modifications of cervical; 53 of these genes showed at least a twofold change in one or more experimental group in relation to onset of estrus, and the differences between groups were significant ( P < 0.05). The majority of these genes showed the greatest alteration in their expression in the 48 h postestrus and luteal phase groups.
Collapse
Affiliation(s)
- Katarzyna Pluta
- Veterinary Sciences Centre, University College Dublin, Belfield, Dublin, Ireland
| | - Paul A. McGettigan
- Veterinary Sciences Centre, University College Dublin, Belfield, Dublin, Ireland
| | - Colm J. Reid
- Veterinary Sciences Centre, University College Dublin, Belfield, Dublin, Ireland
| | - John A. Browne
- Veterinary Sciences Centre, University College Dublin, Belfield, Dublin, Ireland
| | - Jane A. Irwin
- Veterinary Sciences Centre, University College Dublin, Belfield, Dublin, Ireland
| | - Tharmala Tharmalingam
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland; and
| | | | - Alan Baird
- Veterinary Sciences Centre, University College Dublin, Belfield, Dublin, Ireland
| | - Brendan J. Loftus
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland; and
| | - Alexander C. O. Evans
- Veterinary Sciences Centre, University College Dublin, Belfield, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland; and
| | | |
Collapse
|
37
|
Dolan B, Naughton J, Tegtmeyer N, May FEB, Clyne M. The interaction of Helicobacter pylori with the adherent mucus gel layer secreted by polarized HT29-MTX-E12 cells. PLoS One 2012; 7:e47300. [PMID: 23056622 PMCID: PMC3466223 DOI: 10.1371/journal.pone.0047300] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 09/13/2012] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori colonises the gastric mucosa of humans. The majority of organisms live in mucus. These organisms are an important reservoir for infection of the underlying epithelium. Cell culture models for H. pylori infection do not normally possess a mucus layer. The interaction of H. pylori with TFF1, a member of the trefoil factor family found in gastric mucin, is mediated by lipopolysaccharide. To test the hypothesis that the interaction of H. pylori with TFF1 promotes mucus colonization we characterised the interaction of H. pylori with a mucus secreting cell line, HT29-MTX-E12. An isogenic mutant of H. pylori with truncated core oligosaccharides was produced and binding to TFF1 and ability to colonise HT29-MTX-E12 cells determined. The adherent mucus layer of HT29-MTX-E12 cells contained the gastric mucin MUC5AC and trefoil factors, TFF1 and TFF3. H. pylori was found within the mucus layer in discrete clusters and in close association with TFF1. It also interacted with the membrane bound mucin MUC1 and replicated when co-cultured with the cells. An isogenic mutant of H. pylori with a truncated LPS core did not interact with TFF1, and colonization of HT29-MTX-E12 cells was reduced compared to the wild-type strain (p<0.05). Preincubation of cells with wild type LPS but not with truncated LPS resulted in reduced colonization by H. pylori. These results demonstrate that the interaction of TFF1 with H. pylori is important for colonization of gastric mucus and the core oligosaccharide of H. pylori LPS is critical for this interaction to occur. HT29-MTX-E12 cells are a useful system with which to study the interaction of bacteria with mucosal surfaces and the effect of such interactions on mediating colonization.
Collapse
Affiliation(s)
- Brendan Dolan
- University College Dublin, School of Medicine and Medical Science, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Science, Belfield, Dublin, Ireland
- The National Childrens Research Centre, Dublin, Ireland
| | - Julie Naughton
- University College Dublin, School of Medicine and Medical Science, Dublin, Ireland
- School of Biomolecular and Biomedical Science, Dublin, Ireland
| | - Nicole Tegtmeyer
- School of Biomolecular and Biomedical Science, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Science, Belfield, Dublin, Ireland
| | - Felicity E. B. May
- Faculty of Medical Sciences, University of Newcastle upon Tyne, Newcastle upon Tyne, United Kingdom
| | - Marguerite Clyne
- University College Dublin, School of Medicine and Medical Science, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Science, Belfield, Dublin, Ireland
- The National Childrens Research Centre, Dublin, Ireland
- * E-mail:
| |
Collapse
|
38
|
Song JY, Kim BW, Lee AW, Lee KY, Chung IS, Lee BI, Choi H, Ji JS, Chae HS, Choi KY. Expression of MUC5AC and Trefoil Peptide 1 (TFF1) in the Subtypes of Intestinal Metaplasia. Clin Endosc 2012; 45:151-154. [PMID: 22866256 PMCID: PMC3401619 DOI: 10.5946/ce.2012.45.2.151] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 11/23/2011] [Accepted: 11/23/2011] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND/AIMS Alterations of the expression pattern of mucins and trefoil peptides have been described in gastric adenocarcinomas and in their precursor lesions. The aim of this study was to determine the progression patterns of intestinal metaplasia (IM) subtypes by analyzing the expression patterns of TFF1 and MUC5AC in different subtypes of IM of the stomach. METHODS Endoscopic gastric biopsies of the antrum and body were obtained from patients with dyspepsia and endoscopic IM. Alcian blue/periodic acid-Schiff staining and the high iron diamine technique were used to classify the subtypes of IM. Immunoreactivity for MUC5AC and TFF1 was estimated in different types of IM. RESULTS IM was detected in 128 samples from 80 patients; type I was found in 48 samples, type II was found in 37 samples, and type III was found in 43 samples. There was a gradual decrease in MUC5AC and TFF1 expression during the progression of IM from type I to type III via the type II intermediate. CONCLUSIONS This downregulation of MUC5AC and TFF1 expression may challenge the sequential progression of IM from type I to type III via the type II intermediate, and it might be associated with gastric carcinogenesis.
Collapse
Affiliation(s)
- Joo-Yong Song
- Department of Internal Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Byung-Wook Kim
- Department of Internal Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Ah-Won Lee
- Department of Anatomic Pathology, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Kyo-Young Lee
- Department of Anatomic Pathology, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - In-Sik Chung
- Department of Internal Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Bo-In Lee
- Department of Internal Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Hwang Choi
- Department of Internal Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Jeong-Seon Ji
- Department of Internal Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Hiun-Suk Chae
- Department of Internal Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Kyu-Yong Choi
- Department of Internal Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea
| |
Collapse
|
39
|
Kubota S, Yamauchi K, Sugano M, Kawasaki K, Sugiyama A, Matsuzawa K, Akamatsu T, Ohmoto Y, Ota H. Pathophysiological investigation of the gastric surface mucous gel layer of patients with Helicobacter pylori infection by using immunoassays for trefoil factor family 2 and gastric gland mucous cell-type mucin in gastric juice. Dig Dis Sci 2011; 56:3498-506. [PMID: 21559742 DOI: 10.1007/s10620-011-1724-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Accepted: 04/15/2011] [Indexed: 12/29/2022]
Abstract
BACKGROUND The trefoil factor family (TFF) 2 protein is produced by gastric gland mucous cells (GMCs), and the secreted TFF2 shares a mucosal barrier function with GMC-type mucin. Recently, we presented an enzyme-linked immunosorbent assay (ELISA) method for measurement of GMC-type mucin in the gastric juice. AIMS We aimed to develop an ELISA for TFF2 and to assess pathophysiological changes in the gastric surface mucous gel layer (SMGL) of patients with Helicobacter pylori infection. METHODS The distribution of TFF2 and GMC-type mucin in the SMGL was immunohistochemically determined. The ELISA for TFF2 was based on a polyclonal goat antibody. Recombinant TFF2 was employed to prepare the calibrators. TFF2 and GMC-type mucin in the gastric juice in healthy individuals (n = 33) and patients with gastritis (n = 37), gastric ulcer (n = 16), and duodenal ulcer (n = 10) were assayed using ELISA. RESULTS TFF2 and GMC-type mucin were immunohistochemically co-localized in the gastric SMGL and GMCs. The TFF2 levels in the patients were significantly higher than those in the healthy individuals. Further, the TFF2 levels in the H. pylori-positive patients were significantly higher than those in the H. pylori-negative patients, and decreased after the eradication of the infection. GMC-type mucin levels showed a tendency similar to that of TFF2 levels. CONCLUSIONS The upregulation of TFF2 and GMC-type mucin secretion may reflect the response of the gastric mucosa to H. pylori-induced injuries. TFF2 and GMC-type mucin secreted into the SMGL may protect the gastric mucosa against H. pylori.
Collapse
Affiliation(s)
- Seiko Kubota
- Department of Laboratory Medicine, Shinshu University Hospital, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Laboratory mice have become one of the best animal species for mechanistic studies in gastrointestinal research. Their abundant genetic information, the way of causing carcinogenesis easily by transgenic and gene knockout techniques, limited effort in time and costs, and their practicability provide advantages over other animal models. Meanwhile, several murine practical models have been established for the investigation of the initiation, expansion, and progression of gastritis and gastric carcinoma, for assessing the effects of bacterial, genetic and environmental factors, and for evaluating therapeutic and preventive strategies in gastric diseases. This article gives a review of murine models of gastritis and gastric cancer, placing emphasis on the models associated with Helicobacter pylori infection and techniques used in our laboratory. We discuss matters of murine gastric anatomy, as well as techniques of infection, tissue preparation, and histology.
Collapse
|
41
|
McGuckin MA, Lindén SK, Sutton P, Florin TH. Mucin dynamics and enteric pathogens. Nat Rev Microbiol 2011. [PMID: 21407243 DOI: 10.1038/nrm] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The extracellular secreted mucus and the cell surface glycocalyx prevent infection by the vast numbers of microorganisms that live in the healthy gut. Mucin glycoproteins are the major component of these barriers. In this Review, we describe the components of the secreted and cell surface mucosal barriers and the evidence that they form an effective barricade against potential pathogens. However, successful enteric pathogens have evolved strategies to circumvent these barriers. We discuss the interactions between enteric pathogens and mucins, and the mechanisms that these pathogens use to disrupt and avoid mucosal barriers. In addition, we describe dynamic alterations in the mucin barrier that are driven by host innate and adaptive immune responses to infection.
Collapse
Affiliation(s)
- Michael A McGuckin
- Immunity, Infection and Inflammation Program, Mater Medical Research Institute and The University of Queensland School of Medicine, South Brisbane, Queensland 4101, Australia.
| | | | | | | |
Collapse
|
42
|
|
43
|
Schmitz JM, Durham CG, Ho SB, Lorenz RG. Gastric mucus alterations associated with murine Helicobacter infection. J Histochem Cytochem 2009; 57:457-67. [PMID: 19153195 DOI: 10.1369/jhc.2009.952473] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The C57BL/6 mouse has been shown to develop gastric adenocarcinoma after Helicobacter felis infection. This model was used to determine whether mucin and trefoil factor (TFF) expression after infection was altered in a similar fashion to the changes seen in the protective gastric mucus layer of the human stomach after H. pylori infection. Our results indicate that this mouse model mimics many of the changes seen after human H. pylori infection, including increased expression of muc4 and muc5b and loss of muc5ac. These alterations in mucin expression occurred as early as 4 weeks postinfection, before the development of significant mucous metaplasia or gastric dysplasia. The decrease in muc5ac expression occurred only in the body of the stomach and was not secondary to the adaptive immune response to infection, because a similar decrease in expression was seen after infection of B6.Rag-1(-/-) mice, which lack B and T cells. Intriguingly, the increased expression of Muc4 and Muc5b in infected C57BL/6 mice was not seen in the infected B6.Rag-1(-/-) mice. Because B6.Rag-1(-/-) mice do not develop gastric pathology after H. felis infection, these findings point to the potential role of Muc4 and Muc5b in disease progression. This manuscript contains online supplemental material at http://www.jhc.org. Please visit this article online to view these materials.
Collapse
Affiliation(s)
- Julia M Schmitz
- Department of Microbiology, University of Alabama at Birmingham, 1825 University Boulevard, SHEL 602, Birmingham, AL 35294-2182, USA
| | | | | | | |
Collapse
|
44
|
Reeves EP, Ali T, Leonard P, Hearty S, O'Kennedy R, May FEB, Westley BR, Josenhans C, Rust M, Suerbaum S, Smith A, Drumm B, Clyne M. Helicobacter pylori lipopolysaccharide interacts with TFF1 in a pH-dependent manner. Gastroenterology 2008; 135:2043-54, 2054.e1-2. [PMID: 18848942 DOI: 10.1053/j.gastro.2008.08.049] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2008] [Revised: 08/15/2008] [Accepted: 08/22/2008] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Little is known about how bacteria establish chronic infections of mucosal surfaces. Helicobacter pylori (H. pylori), a chronic pathogen that lives in the gastric mucosa of humans, interacts with the trefoil factor family (TFF) protein TFF1, which is found in gastric mucus. We aimed to characterize the interaction of H. pylori with TFF1 and to assess the role of this interaction in mediating colonization. METHODS Subcellular fractions of H. pylori were immobilized and then probed with TFF1, TFF2, or TFF3. The effect of glycosidases and preincubation with monosaccharides on the interaction and binding of TFF1 to a H. pylori adhesin was assessed. The interaction between H. pylori adhesin and TFF1 was characterized using surface plasmon resonance, flow cytometry, nondenaturing polyacrylamide gel electrophoresis, coimmunofluoresence, and incubation with tissue sections. RESULTS The H. pylori core oligosaccharide portion (rough form) of lipopolysaccharide (RF-LPS) bound to TFF1 and to a lesser extent TFF3; this interaction was inhibited by incubation of RF-LPS with mannosidase, glucosidase, or mixed monosaccharides. TFF1 also bound to human serum albumin-conjugated mannose and glucose. The optimum pH for binding was 5.0-6.0 for TFF1 and 7.0 for TFF3. H. pylori bound TFF1 in gastric mucus ex vivo; binding of LPS-coated latex beads to human antral gastric tissue was inhibited by TFF1. CONCLUSIONS TFF1 interacts specifically with H. pylori RF-LPS. The pH dependence of this interaction indicates that binding of H. pylori to TFF1 in the stomach could promote colonization of the mucus layer adjacent to the gastric epithelial surface.
Collapse
Affiliation(s)
- Emer P Reeves
- University College Dublin School of Medicine and Medical Science, The Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
The trefoil factor interacting protein TFIZ1 binds the trefoil protein TFF1 preferentially in normal gastric mucosal cells but the co-expression of these proteins is deregulated in gastric cancer. Int J Biochem Cell Biol 2008; 41:632-40. [PMID: 18722547 PMCID: PMC2632736 DOI: 10.1016/j.biocel.2008.07.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 07/17/2008] [Accepted: 07/18/2008] [Indexed: 01/15/2023]
Abstract
The gastric tumour suppressor trefoil protein TFF1 is present as a covalently bound heterodimer with a previously uncharacterised protein, TFIZ1, in normal human gastric mucosa. The purpose of this research was firstly to examine the molecular forms of TFIZ1 present, secondly to determine if TFIZ1 binds other proteins apart form TFF1 in vivo, thirdly to investigate if TFIZ1 and TFF1 are co-regulated in normal gastric mucosa and fourthly to determine if their co-regulation is maintained or disrupted in gastric cancer. We demonstrate that almost all human TFIZ1 is present as a heterodimer with TFF1 and that TFIZ1 is not bound to either of the other two trefoil proteins, TFF2 and TFF3. TFIZ1 and TFF1 are co-expressed by the surface mucus secretory cells throughout the stomach and the molecular forms of each protein are affected by the relative abundance of the other. TFIZ1 expression is lost consistently, early and permanently in gastric tumour cells. In contrast, TFF1 is sometimes expressed in the absence of TFIZ1 in gastric cancer cells and this expression is associated with metastasis (lymph node involvement: p = 0.007). In conclusion, formation of the heterodimer between TFIZ1 and TFF1 is a specific interaction that occurs uniquely in the mucus secretory cells of the stomach, co-expression of the two proteins is disrupted in gastric cancer and expression of TFF1 in the absence of TFIZ1 is associated with a more invasive and metastatic phenotype. This indicates that TFF1 expression in the absence of TFIZ1 expression has potentially deleterious consequences in gastric cancer.
Collapse
|
46
|
Thornton DJ, Rousseau K, McGuckin MA. Structure and function of the polymeric mucins in airways mucus. Annu Rev Physiol 2008; 70:459-86. [PMID: 17850213 DOI: 10.1146/annurev.physiol.70.113006.100702] [Citation(s) in RCA: 616] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The airways mucus gel performs a critical function in defending the respiratory tract against pathogenic and environmental challenges. In normal physiology, the secreted mucins, in particular the polymeric mucins MUC5AC and MUC5B, provide the organizing framework of the airways mucus gel and are major contributors to its rheological properties. However, overproduction of mucins is an important factor in the morbidity and mortality of chronic airways disease (e.g., asthma, cystic fibrosis, and chronic obstructive pulmonary disease). The roles of these enormous, multifunctional, O-linked glycoproteins in health and disease are discussed.
Collapse
Affiliation(s)
- David J Thornton
- Wellcome Trust Center for Cell Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom.
| | | | | |
Collapse
|
47
|
Thuwajit P, Chawengrattanachot W, Thuwajit C, Sripa B, Paupairoj A, Chau-In S. Enhanced expression of mucin 6 glycoprotein in cholangiocarcinoma tissue from patients in Thailand as a prognostic marker for survival. J Gastroenterol Hepatol 2008; 23:771-8. [PMID: 18410610 DOI: 10.1111/j.1440-1746.2008.05332.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIM Cholangiocarcinoma (CCA) is a mucin-producing cancer that has poor prognosis. Mucin 6 (MUC6) is a mucin that is normally co-expressed with the trefoil factor family-2 (TFF2) trefoil peptide. Both MUC6 and TFF2 have been reported to be involved in the progression of many types of cancers. The aim of this study was to determine the expression of MUC6 and TFF2 in CCA tissues and associate these results with clinical data. METHODS MUC6 and TFF2 were detected in CCA tissues by immunohistochemistry. The correlations of MUC6 and TFF2 expressions with clinical data were analyzed. RESULTS We determined the significant co-expression of both proteins in serial CCA tissues. The high expressions of MUC6 and TFF2 were demonstrated in 37% and 31% of patients, respectively. The expression levels decreased in the advanced stage of CCA when clinical metastasis was exhibited. The high expression of either protein showed a correlation with prolonged postoperative survival time, but only a high expression of MUC6 is significantly correlated with a 5-year survival rate. A multivariate Cox regression analysis revealed that a low expression of MUC6, high expression of TFF2, age of patients >56 years, tumor size >5 cm, and poorly-differentiated histological type were independent, poor prognostic indicators for CCA. CONCLUSION MUC6 showed a good correlation with the survival of CCA patients. It may be of value to propose that MUC6 is a good prognostic marker for CCA management.
Collapse
Affiliation(s)
- Peti Thuwajit
- Department of Biochemistry, and Liver Fluke and Cholabngiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.
| | | | | | | | | | | |
Collapse
|
48
|
Ichikawa T, Ota H, Sugiyama A, Maruta F, Ikezawa T, Hotta K, Ishihara K. Effects of a novel histamine H2-receptor antagonist, lafutidine, on the mucus barrier of human gastric mucosa. J Gastroenterol Hepatol 2007; 22:1800-5. [PMID: 17914953 DOI: 10.1111/j.1440-1746.2006.04721.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM Lafutidine is a novel histamine H(2)-receptor antagonist used primarily as an antisecretory agent in Japan. Previous human studies have not assessed its gastroprotective effects. The purpose of the present study was to determine the effects of lafutidine on the human gastric mucus layer using both histological and biochemical methods. METHODS Of the 14 patients scheduled for gastrectomy who consented to participate, seven were given 14 days of lafutidine 20 mg/day (lafutidine group) and the others received no medication (control group). The surface mucus gel layer in Carnoy-fixed tissue sections was examined immunohistochemically. Both the thickness of the mucus layer and its mucin content were measured in gastric corpus mucosa. RESULTS There was no detectable difference between the groups in the grade of gastritis or the immunohistochemical staining characteristics. The laminated structure of the surface mucus gel layer was retained after administration of lafutidine and it was thicker than the layer in the control group. The surface layer in the lafutidine group had three-fold more mucin than that in the control group. There was no difference between the two groups in the mucin content of the deep mucosa. CONCLUSION Lafutidine, given at clinical dosages, not only inhibits acid secretion but also strengthens the mucus barrier of the human gastric mucosa.
Collapse
Affiliation(s)
- Takafumi Ichikawa
- Department of Biochemistry, Kitasato University School of Medicine, Sagamihara, Japan.
| | | | | | | | | | | | | |
Collapse
|
49
|
Kouznetsova I, Laubinger W, Kalbacher H, Kalinski T, Meyer F, Roessner A, Hoffmann W. Biosynthesis of Gastrokine-2 in the Human Gastric Mucosa: Restricted Spatial Expression along the Antral Gland Axis and Differential Interaction with TFF1, TFF2 and Mucins. Cell Physiol Biochem 2007; 20:899-908. [DOI: 10.1159/000110450] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2007] [Indexed: 01/07/2023] Open
|
50
|
Altered expression of CDX-2, PDX-1 and mucin core proteins in "Ulcer-associated cell lineage (UACL)" in Crohn's disease. J Mol Histol 2007; 39:161-8. [PMID: 17957487 DOI: 10.1007/s10735-007-9149-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Accepted: 09/27/2007] [Indexed: 12/15/2022]
Abstract
The ulcer-associated cell lineage (UACL) induced at the site of ileac chronic ulceration in Crohn's disease has been reported to show histological differentiation resembling gastric pyloric mucosa. To clarify the significance of homeobox gene-encoded transcription factors in the formation of the UACL in Crohn's disease, we investigated the immunohistochemical expression of gastrointestinal mucins (MUC5AC for gastric surface mucous cells; MUC6 for gastric gland mucous cells, and MUC2 for intestinal goblet cells) and homeobox gene-encoded transcription factors (CDX-2 for intestinal mucosa and PDX-1 for pyloric mucosa) in the UACL. The analysis was undertaken on ileal mucosa obtained from ileal resections performed in 19 patients with active Crohn's disease of the small bowel. The UACL was observed in nine patients. In the UACL, expression of mucous cells with a foveolar-structure showed immunoreactivity to MUC5AC, and the mucous cells with a glandular structure showed immunoreactivity to MUC6, and the expression of MUC2 was decreased. In addition, we detected the decreased expression of CDX-2 along with the increased expression of PDX-1 in the UACL. The UACL showed histological differentiation simulating gastric pylori mucosa. The down-regulation of CDX-2 and the up-regulation of PDX-1 could be an important mechanism in the induction of the UACL.
Collapse
|