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
|
Immunoexpression of Trefoil Factor 1 in Non-Neoplastic and Neoplastic Canine Gastric Tissues. Animals (Basel) 2021; 11:ani11102855. [PMID: 34679875 PMCID: PMC8532865 DOI: 10.3390/ani11102855] [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: 08/18/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary Gastric carcinoma (GC) is the second leading cause of death in humans and the most frequent malignancy in the stomach of dogs. As in humans, the prognosis of canine gastric cancer is generally poor owing to the advanced stage at the time of diagnosis, resulting in limited treatment options. In dogs, the molecular mechanisms involved in the growth and progression of gastric cancer remain largely unknown. Trefoil factor 1 (TFF1) protein is a mucin-associated secretory molecule that plays an important role in the maintenance and protection of epithelial surface integrity. Some human studies showed that TFF1 can protect mucosa against damage and suppress carcinogenesis, while other studies showed that TFF1 can restrict cell adhesion, promote tumor cell invasion, and block necrosis of tumor cells. In human gastric cancer, TFF1 has been found to decrease, and it has been proposed that it might act as a tumor suppressor factor. The present study was carried out to investigate whether there is a relationship between TFF1 and canine gastric carcinogenesis. We found an association between reduced expression of TFF1 and the development and progression of gastric cancer in dogs. The pathological and behavioral similarities between spontaneous canine GC and human counterparts make it logical to assume that dogs may be a useful model for human gastric cancer. Abstract TFF1 expression is markedly reduced in human GCs, suggesting that TFF1 is a tumor suppressor for human gastric cancer. The present study evaluated the expression and distribution pattern of TFF1 in paraffin-embedded canine gastric tissue samples, including normal mucosa (n = 3), polyps (n = 8), carcinomas (n = 31) and their adjacent non-neoplastic mucosa (n = 30), neoplastic emboli (n = 14), and metastatic lesions (n = 9), by immunohistochemistry (IHC). All normal gastric tissues expressed TFF1 in the superficial foveolar epithelium and mucopeptic cells of the neck region. Most gastric polyps (GPs) displayed immunoreactivity for TFF1 in >75% of the epithelial component. In GCs, the expression of TFF1 was found reduced in 74.2% of the cases. The level of TFF1 expression had a decreased tendency from normal gastric mucosa to GPs and GCs (p < 0.05). No significant differences in the expression of TFF1 were found in GCs, according to age, sex, histological type based on World Health Organization (WHO) and Lauren classification, tumor location, depth of tumor invasion, presence of neoplastic emboli or metastatic lesions. The median survival time of GC patients with preserved and reduced TFF1 immunoexpression were 30 and 12 days, respectively. Kaplan–Meier analysis revealed no significant survival differences between the two groups (p > 0.05). These findings suggest that TFF1 protein may play a role in canine gastric carcinogenesis, and further studies are necessary to define its usefulness as a prognostic indicator in canine gastric carcinoma.
Collapse
|
3
|
Novel urinary protein biomarker panel for early diagnosis of gastric cancer. Br J Cancer 2020; 123:1656-1664. [PMID: 32934343 PMCID: PMC7686371 DOI: 10.1038/s41416-020-01063-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 08/19/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022] Open
Abstract
Background With the goal of discovering non-invasive biomarkers for early diagnosis of GC, we conducted a case-control study utilising urine samples from individuals with predominantly early GC vs. healthy control (HC). Methods Among urine samples from 372 patients, age- and sex-matched 282 patients were randomly divided into three groups: 18 patients in a discovery cohort; 176 patients in a training cohort and 88 patients in a validation cohort. Results Among urinary proteins identified in the comprehensive quantitative proteomics analysis, urinary levels of TFF1 (uTFF1) and ADAM12 (uADAM12) were significantly independent diagnostic biomarkers for GC, in addition to Helicobacter pylori status. A urinary biomarker panel combining uTFF1, uADAM12 and H. pylori significantly distinguished between HC and GC patients in both training and validation cohorts. On the analysis for sex-specific biomarkers, this combination panel demonstrated a good AUC of 0.858 for male GC, whereas another combination panel of uTFF1, uBARD1 and H. pylori also provided a good AUC of 0.893 for female GC. Notably, each panel could distinguish even stage I GC patients from HC patients (AUC = 0.850 for males; AUC = 0.845 for females). Conclusions Novel urinary protein biomarker panels represent promising non-invasive biomarkers for GC, including early-stage disease.
Collapse
|
4
|
Suárez C, Vizoso F, Rodríguez JC, García I, Raigoso P, Allende MT, García-Muñíz JL, García-Morán M. Prognostic Significance of Cytosolic pS2 Protein Content in Gastric Cancer. Int J Biol Markers 2018; 16:37-44. [PMID: 11288953 DOI: 10.1177/172460080101600105] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
pS2, a 60-amino-acid chain peptide which is the most widespread estrogen-induced RNA messenger in MCF-7 breast cancer cells, is normally detected in the epithelium of gastric mucosa. The aims of this work were to evaluate the cytosolic pS2 content and its clinical significance in gastric carcinomas. Cytosolic pS2 levels were examined by immunoradiometric methods in 108 patients with primary gastric adenocarcinomas. The mean follow-up period was 23.3 months. The cytosolic pS2 levels of the tumors ranged widely, i.e., from 0.1 to 3217 ng/mg protein. There were no significant differences in pS2 content between tumors (mean ± standard error: 137.2±31.4 ng/mg protein) and paired adjacent mucosa samples (n=84; mean ± standard error: 249.6±32.6 ng/mg protein), nor were there any significant differences in tumoral pS2 levels with respect to clinicopathologic parameters such as patient age and sex or tumor location, stage, histologic type or grade. However, the results indicated that high intratumoral pS2 levels were significantly and independently associated with an unfavorable outcome in the overall group of patients (p=0.0266) and in patients with resectable gastric cancer (p=0.003). In conclusion, pS2 may represent a useful biological marker in gastric cancer.
Collapse
Affiliation(s)
- C Suárez
- Department of General Surgery, Hospital de Jove, Gijón, Spain
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Vizoso FJ, Fagilde MC, Corte MD, Corte MG, Gava R, Bongera M, Allende MT, García-Muñiz JL. Cytosolic Levels of An Estrogen-Induced Breast Cancer-Associated Peptide (TFF1/pS2) in Colorectal Cancer: Clinical Significance and Relationship with Steroid Receptors. Int J Biol Markers 2018; 18:301-10. [PMID: 14756547 DOI: 10.1177/172460080301800409] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background The Trefoil Factor 1 (TFF1/pS2), a peptide consisting of 60 amino acids, is the most abundant estrogen-induced messenger RNA in MCF-7 breast cancer cells and is also expressed by colorectal carcinomas. The objective of this work was to evaluate the cytosolic TFF1 content in colorectal carcinomas, its possible relationship with estrogen and progesterone receptors as well as with clinicopathological tumor parameters, and its potential prognostic significance. Methods Cytosolic TFF1 levels were examined by immunoradiometric assay in 178 patients with resectable colorectal cancer. The mean follow-up period was 32 months. Results There was a wide variability of cytosolic TFF1 levels in tumor-surrounding mucosa samples (0.09-42.5 ng/mg protein) as well as in tumors (0.01-270 ng/mg protein). Comparison of paired mucosa and carcinoma samples showed significantly higher TFF1 levels in tumors (mean: 17.1 ng/mg protein) than in mucosa samples (10 ng/mg protein) (p=0.027). TFF1 levels were significantly higher in mucosa samples surrounding distal colon and rectal tumors (p=0.0001) and in tumor samples obtained from older patients (p=0.007). However, there were no significant differences in tumor TFF1 levels with respect to clinicopathological parameters such as the patient's sex, tumor location, stage, histological grade, ploidy, S-phase, or tumor estrogen and progesterone receptors. In addition, there was no significant relationship between tumor TFF1 levels and disease outcome. Conclusions TFF1 may play an as yet undetermined role in the tumorigenesis of colorectal carcinomas. However, cytosolic levels of TFF1 do not seem to have any prognostic significance in colorectal carcinomas.
Collapse
Affiliation(s)
- F J Vizoso
- Servicio de Cirugía General, Hospital de Jove, , Asturias, Gijón, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
6
|
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: 0.9] [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
|
7
|
Huang Z, Zhang X, Lu H, Wu L, Wang D, Zhang Q, Ding H. Serum trefoil factor 3 is a promising non-invasive biomarker for gastric cancer screening: a monocentric cohort study in China. BMC Gastroenterol 2014; 14:74. [PMID: 24720760 PMCID: PMC4012276 DOI: 10.1186/1471-230x-14-74] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 03/24/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The search for better non-invasive biomarkers for gastric cancer remains ongoing. We investigated the predictive power of serum trefoil factor (TFF) levels as biomarkers for gastric cancer in comparison with the pepsinogen (PG) test. METHODS Patients with gastric cancer, chronic atrophic gastritis (CAG) or chronic non-atrophic gastritis (CNAG), and healthy people were recruited. Serum concentrations of TFFs, PG I, and PG II, as well as the presence of antibodies against Helicobacter pylori, were measured by enzyme-linked immunosorbent assays (ELISA). Receiver operating characteristics (ROC) were used to compare the predictive powers of the selected factors. RESULTS The serum concentrations of TFF1, TFF2, and TFF3 in the control groups were significantly lower than those in the gastric cancer group with the exception of TFF2 which was elevated in CAG. The area under the ROC curve for TFF3 was greater than that for the PG I/II ratio (0.81 vs 0.78). TFF3 also had a significantly higher predictive power for distinguishing gastric cancer than the PG test (odds ratio: 10.33 vs 2.57). Moreover, combining the serum TFF3 and PG tests for gastric cancer had better predictive power than either alone. CONCLUSIONS Serum TFF3 may be a better predictor of gastric cancer than the PG test, while the combined testing of serum PG and TFF3 could further improve the efficacy of gastric cancer screening.
Collapse
Affiliation(s)
- Zhigang Huang
- Department of Gastroenterology, Lihuili Hospital of Ningbo Medical Center, 57# Xingning Road, Ningbo 315000, China.
| | | | | | | | | | | | | |
Collapse
|
8
|
Im S, Yoo C, Jung JH, Choi HJ, Yoo J, Kang CS. Reduced expression of TFF1 and increased expression of TFF3 in gastric cancer: correlation with clinicopathological parameters and prognosis. Int J Med Sci 2013; 10:133-40. [PMID: 23329884 PMCID: PMC3547210 DOI: 10.7150/ijms.5500] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 12/26/2012] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES The trefoil factor family (TFF) is composed of three thermostable, and protease-resistant proteins, named TFF1, TFF2 and TFF3, and plays a role in gastrointestinal mucosal defence and repair. Recently, TFFs have been found to be related to the development of various types of cancer. This study assessed the relationship between the expression of TFF1 and TFF3 and the clinicopathological parameters in gastric carcinoma (GC). MATERIALS AND METHODS The expression of TFF1 and TFF3 was analyzed by immunohistochemistry in 292 GCs and 20 normal gastric tissues. RESULTS All normal gastric tissues expressed TFF1, but 53.8% of GCs showed reduced TFF1 expression. However, TFF3 was not detected in normal gastric tissues and 44.2% of GCs showed a high level of expression. Highly expressed TFF3 was significantly correlated with lymph node metastasis, lymphatic invasion, vein invasion, and advanced stage. The overall survival was shorter in patients with high expression of TFF3 than in those with low expression of TFF3 in 292 GCs and in 125 early GCs (EGCs). Moreover, in patients with EGCs, high expression of TFF3, associated with reduced expression of TFF1, was determined as an independent poor prognostic marker. CONCLUSIONS Reduced expression of TFF1 and increased expression of TFF3 may play a role in the carcinogenesis of gastric cancer. Furthermore, high expression of TFF3 with reduced expression of TFF1 may be a marker of poor prognosis for patients with EGC.
Collapse
Affiliation(s)
- Soyoung Im
- Department of Hospital Pathology, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Republic of Korea
| | | | | | | | | | | |
Collapse
|
9
|
Aikou S, Ohmoto Y, Gunji T, Matsuhashi N, Ohtsu H, Miura H, Kubota K, Yamagata Y, Seto Y, Nakajima A, Goldenring JR, Kaminishi M, Nomura S. Tests for serum levels of trefoil factor family proteins can improve gastric cancer screening. Gastroenterology 2011; 141:837-845.e1-7. [PMID: 21699780 PMCID: PMC3163741 DOI: 10.1053/j.gastro.2011.05.040] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 05/12/2011] [Accepted: 05/20/2011] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Improving methods for early detection of gastric cancer could reduce mortality. Measurements of serum pepsinogen levels have been used for screening in Japan without satisfactory levels of sensitivity or specificity. Trefoil factor family (TFF) proteins (TFF1, TFF2, and TFF3) are small and stable molecules secreted by the mammalian gastrointestinal tract. Foveolar hyperplasia, spasmolytic polypeptide (TFF2)-expressing metaplasia, and intestinal metaplasia are histologic changes observed in patients with atrophic gastritis; they express TFF1, TFF2, and TFF3, respectively. We investigated whether serum levels of TFF can be used as markers for gastric cancer screening. METHODS Serum was collected from 183 patients with gastric cancer and 280 healthy individuals without cancer. Serum levels of anti-Helicobacter pylori immunoglobulin G, pepsinogen I, pepsinogen II, TFF1, TFF2, and TFF3 were measured by enzyme-linked immunosorbent assay and associated with gastric cancer. RESULTS Using a cutoff of 3.6 ng/mL, the level of TFF3 was significantly increased in serum samples from patients with cancer (odds ratio, 18.1; 95% confidence interval, 11.2-29.2); using this test, patients with cancer were identified with 80.9% sensitivity and 81.0% specificity. The test for TFF3 had a significantly higher odds ratio than that for pepsinogen. A test for the combination of TFF3 and pepsinogen had better results than the test for only pepsinogen. CONCLUSIONS Serum levels of TFF3 are a better marker of gastric cancer than pepsinogen; a test for the combined levels of serum pepsinogen and TFF3 could improve gastric cancer screening.
Collapse
Affiliation(s)
- Susumu Aikou
- Department of Gastrointestinal Surgery, Graduate School of Medicine, University of Tokyo
| | | | | | | | - Hiroshi Ohtsu
- Department of Clinical Trial Data Management, Graduate School of Medicine, University of Tokyo
| | - Hirona Miura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, University of Tokyo
| | - Kensuke Kubota
- Department of Gastroenterology, School of Medicine, Yokohama City University
| | - Yukinori Yamagata
- Department of Gastrointestinal Surgery, Graduate School of Medicine, University of Tokyo
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, University of Tokyo
| | - Atsushi Nakajima
- Department of Gastroenterology, School of Medicine, Yokohama City University
| | - James R Goldenring
- Nashville VA Medical Center and the Departments of Surgery and Cell and Developmental Biology, Epithelial Biology Center, Vanderbilt University School of Medicine
| | | | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, University of Tokyo
| |
Collapse
|
10
|
Affiliation(s)
- Won Sang Park
- Department of Pathology, The Catholic University of Korea, School of Medicine,
| |
Collapse
|
11
|
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: 1.2] [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
|
12
|
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: 2.0] [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
|
13
|
Moss SF, Lee JW, Sabo E, Rubin AK, Rommel J, Westley BR, May FEB, Gao J, Meitner PA, Tavares R, Resnick MB. Decreased expression of gastrokine 1 and the trefoil factor interacting protein TFIZ1/GKN2 in gastric cancer: influence of tumor histology and relationship to prognosis. Clin Cancer Res 2008; 14:4161-7. [PMID: 18593995 DOI: 10.1158/1078-0432.ccr-07-4381] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE Transcriptional profiling showed decreased expression of gastrokine 1 (GKN1) and the related trefoil factor interacting protein (TFIZ1/GKN2) in Helicobacter pylori infection. Decreased GKN1 and GKN2 mRNA expression has been reported in gastric adenocarcinoma. We have examined GKN1 and GKN2 protein expression in a large gastric cancer series, correlated expression with tumor subtype, and evaluated their utility as prognostic biomarkers. EXPERIMENTAL DESIGN GKN1, GKN2, and the trefoil factors TFF1 and TFF3 were examined in tissue microarrays from 155 distal gastric adenocarcinomas. Immunohistochemical expression was correlated with clinical outcome. GKN1 and GKN2 expression was measured by real-time PCR and Western analysis in samples of gastric cancer and adjacent nonneoplastic mucosa. RESULTS GKN1 was lost in 78% of diffuse and 42% of intestinal cancers (P < 0.0001, diffuse versus intestinal). GKN2 expression was lost in 85% of diffuse and 54% of intestinal type cancers (P < 0.002). GKN1 and GKN2 down-regulation were confirmed by Western and real-time PCR analysis. Loss of either protein was associated with significantly worse outcome in intestinal-type tumors by univariate analysis; and GKN2 loss remained a predictor of poor outcome in multivariate analysis (P < 0.033). TFF1 was lost in >70%, and TFF3 was expressed in approximately 50% of gastric cancers. CONCLUSIONS Loss of GKN1 and GKN2 expression occurs frequently in gastric adenocarcinomas, especially in the diffuse subtype. GKN1 and GKN2 loss are associated with shorter overall survival in the intestinal subtype.
Collapse
Affiliation(s)
- Steven F Moss
- Department of Medicine, Gastroenterology Division, Rhode Island Hospital, 593 Eddy Street, APC 414, Providence, RI 02903, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Stomach. Oncology 2007. [DOI: 10.1007/0-387-31056-8_41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
15
|
Green KA, Carroll JS. Oestrogen-receptor-mediated transcription and the influence of co-factors and chromatin state. Nat Rev Cancer 2007; 7:713-22. [PMID: 17721435 DOI: 10.1038/nrc2211] [Citation(s) in RCA: 171] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oestrogen receptor-alpha (ERalpha)-regulated transcription in breast cancer cells involves protein co-factors that contribute to the regulation of chromatin structure. These include co-factors with the potential to regulate histone modifications such as acetylation or methylation, and therefore the transcriptional state of target genes. Although much of the information regarding the interaction of specific co-factors with ER has been generated by studying specific promoter regions, we now have an improved understanding of the nature of these interactions and are better placed to relate these with ER activity and potentially with the activity of breast cancer drugs, including tamoxifen.
Collapse
Affiliation(s)
- Kelly A Green
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, UK
| | | |
Collapse
|
16
|
Thuwajit P, Chawengrattanachot W, Thuwajit C, Sripa B, May FEB, Westley BR, Tepsiri NN, Paupairoj A, Chau-In S. Increased TFF1 trefoil protein expression in Opisthorchis viverrini-associated cholangiocarcinoma is important for invasive promotion. Hepatol Res 2007; 37:295-304. [PMID: 17397518 DOI: 10.1111/j.1872-034x.2007.00045.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AIMS Cholangiocarcinoma (CCA) is a poor prognosis cancer that presents with metastatic disease. This cancer expresses MUC5AC, a mucin which normally co-expresses with trefoil factor family 1 (TFF1) protein. TFF1 is a signalling protein that can activate epithelial cell invasion and has been considered as a metastasis stimulating agent. The aim of this study was to determine the co-expression of TFF1 and MUC5AC in CCA tissues and examine the activity of TFF1 for stimulating the invasive property of CCA cell lines. METHODS In this study, TFF1 and MUC5AC were detected in CCA tissues by using immunohistochemistry. The correlations of both proteins expression with clinical data were analyzed. The activity of TFF1 was investigated using an in vitro invasion assay with established CCA cell lines KKU-100 and KKU-M213. RESULTS We demonstrated a high level of expression of TFF1 in 91.80% of CCA that is associated with a high level of co-expression with MUC5AC in 80.33% of cases. In vitro invasion assay showed that both cell lines have similar responses to TFF1 that could act as both a chemokinetic and chemotactic agent. The dose-response curves were bell-shaped. CONCLUSION TFF1 showed co-expression with MUC5AC in CCA tissues and invasive stimulating activity in vitro. These results may indicate a role for TFF1 in promoting tumor invasion in CCA.
Collapse
Affiliation(s)
- Peti Thuwajit
- Department of Biochemistry, Faculty of Medicine, and Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Xu XQ, Emerald BS, Goh ELK, Kannan N, Miller LD, Gluckman PD, Liu ET, Lobie PE. Gene Expression Profiling to Identify Oncogenic Determinants of Autocrine Human Growth Hormone in Human Mammary Carcinoma. J Biol Chem 2005; 280:23987-4003. [PMID: 15845533 DOI: 10.1074/jbc.m503869200] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have exploited a discrepancy in the oncogenic potential of autocrine and exogenous human growth hormone (hGH) in an attempt to identify molecules that could potentially be involved in oncogenic transformation of the human mammary epithelial cell. Microarray analysis of 19,000 human genes identified a subset of 305 genes in a human mammary carcinoma cell line that were remarkably different in their response to autocrine and exogenous hGH. Autocrine and exogenous hGH also regulated 167 common genes. Semiquantitative reverse transcription-PCR confirmed differential regulation of genes by either autocrine or exogenous hGH. Functional analysis of one of the identified autocrine hGH-regulated genes, TFF3, determined that its expression is sufficient to support anchorage-independent growth of human mammary carcinoma cells. Small interfering RNA-mediated knockdown of TFF3 concordantly abrogated anchorage-independent growth of mammary carcinoma cells and abrogated the ability of autocrine hGH to stimulate oncogenic transformation of immortalized human mammary epithelial cells. Further functional characterization of the identified subset of specifically autocrine hGH regulated genes will delineate additional novel oncogenes for the human mammary epithelial cell.
Collapse
Affiliation(s)
- Xiu Qin Xu
- Microarray and Expression Genomics, Genome Institute of Singapore, Republic of Singapore
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Westley BR, Griffin SM, May FEB. Interaction between TFF1, a Gastric Tumor Suppressor Trefoil Protein, and TFIZ1, a Brichos Domain-Containing Protein with Homology to SP-C. Biochemistry 2005; 44:7967-75. [PMID: 15924415 DOI: 10.1021/bi047287n] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
TFF1 is a gastric tumor suppressor that protects gastric epithelial cells from damage but can promote invasive properties of tumor cells. Antibodies were raised against correctly folded TFF1 protein. These showed that the 6.67 kDa secreted trefoil protein is present as an approximately 25 kDa complex in normal human gastric mucosa. The TFF1 complex was immunopurified from human gastric mucosa and shown to comprise two proteins joined by a disulfide bond. Both were identified by amino-terminal sequencing and MALDI TOF mass spectrometry. The TFF1 protein partner is a previously unknown protein that we have called TFIZ1 for trefoil factor interactions(z) 1. TFIZ1 is expressed and secreted in normal gastric mucosa. TFIZ1 mRNA was cloned from gastric mucosa and sequenced. TFIZ1 is an 18.31 kDa protein and contains an approximately 100 amino acid brichos domain and homology with smart00019.10, SF_P. This is the first demonstration that a member of the trefoil factor family of proteins is bound covalently to a brichos domain-containing protein. The apparent molecular mass of the TFF1:TFIZ1 heterodimer is remarkably close to the theoretical molecular mass of 24.98 kDa. In conclusion, the heterodimer comprises one molecule each of TFF1 and TFIZ1, and the disulfide bond between TFF1 and TFIZ1 is the most important factor stabilizing the heterodimer.
Collapse
Affiliation(s)
- Bruce R Westley
- Department of Pathology, University of Newcastle upon Tyne, UK
| | | | | |
Collapse
|
19
|
Kornprat P, Rehak P, Lemmerer M, Gogg-Kamerer M, Langner C. Analysis of trefoil factor family protein 1 (TFF1, pS2) expression in chronic cholecystitis and gallbladder carcinoma. Virchows Arch 2005; 446:505-10. [PMID: 15821928 DOI: 10.1007/s00428-005-1240-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2004] [Accepted: 02/22/2005] [Indexed: 11/26/2022]
Abstract
Trefoil factor family protein 1 (TFF1, pS2) interacts with mucins to protect gastrointestinal epithelium against injury and contributes to mucosal repair by promoting epithelial cell migration and restitution. Moreover, TFF1 has antiproliferative and anti-apoptotic effects and promotes cell scattering and invasion. We investigated TFF1 expression in healthy and inflamed non-neoplastic gallbladder mucosa as well as in gallbladder carcinomas (n=57) and corresponding metastases (n=18), using a tissue microarray technique. TFF1 immunoreactivity was absent in healthy mucosa, focally observed in epithelium with inflammatory changes and present in 35% of primary and 24% of metastatic cancer tissues. Immunoreactivity significantly decreased with increasing tumour stage (P=0.009) and increasing tumour grade (P=0.001). Patients with TFF1 positive tumours showed a more favourable outcome compared to patients with TFF1 negative tumours in univariate analysis (P=0.006). However, multivariate analysis proved resection status and tumour grade as the only independent prognostic factors. In conclusion, TFF1 is expressed in inflamed non-neoplastic gallbladder epithelium and in low stage and low grade gallbladder carcinomas. Thus, TFF1 may be the missing link between gallstones, chronic cholecystitis and gallbladder cancer. Further studies are needed to evaluate whether TFF1 immunostaining can be used as a diagnostic tool to identify patients with a more favourable outcome.
Collapse
MESH Headings
- Adenocarcinoma/chemistry
- Adenocarcinoma/pathology
- Adenocarcinoma, Clear Cell/chemistry
- Adenocarcinoma, Clear Cell/pathology
- Adenocarcinoma, Mucinous/chemistry
- Adenocarcinoma, Mucinous/pathology
- Adult
- Aged
- Aged, 80 and over
- Analysis of Variance
- Carcinoma/chemistry
- Carcinoma/mortality
- Carcinoma/pathology
- Carcinoma, Adenosquamous/chemistry
- Carcinoma, Adenosquamous/pathology
- Carcinoma, Papillary/chemistry
- Carcinoma, Papillary/pathology
- Carcinoma, Signet Ring Cell/chemistry
- Carcinoma, Signet Ring Cell/pathology
- Cholecystitis/metabolism
- Chronic Disease
- Female
- Gallbladder Neoplasms/chemistry
- Gallbladder Neoplasms/mortality
- Gallbladder Neoplasms/pathology
- Humans
- Immunohistochemistry
- Male
- Middle Aged
- Mucous Membrane/chemistry
- Neoplasm Metastasis
- Neoplasm Staging
- Prognosis
- Proteins/analysis
- Survival Rate
- Trefoil Factor-1
- Tumor Suppressor Proteins
Collapse
Affiliation(s)
- Peter Kornprat
- Department of Surgery, Medical University of Graz, Auenbruggerplatz 29, 8036, Graz, Austria
| | | | | | | | | |
Collapse
|
20
|
Chan MWY, Chan VYW, Leung WK, Chan KK, To KF, Sung JJY, Chan FKL. Anti-sense trefoil factor family-3 (intestinal trefoil factor) inhibits cell growth and induces chemosensitivity to adriamycin in human gastric cancer cells. Life Sci 2005; 76:2581-92. [PMID: 15769482 DOI: 10.1016/j.lfs.2004.11.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2004] [Accepted: 11/11/2004] [Indexed: 01/11/2023]
Abstract
Intestinal trefoil factor (ITF), which is normally absent in gastric mucosa, is over-expressed in gastric cancer. However, the functional significance of ITF in gastric cancer is unknown. We examined the effects of blocking ITF expression on the growth of gastric cancer cells and their responses to chemotherapeutic agents. Anti-sense ITF cDNA was cloned into mammalian expression vector pcDNA3 and was transfected into an ITF-expressing gastric cancer cell line SNU-1. We assessed the doubling time and anchorage dependent growth of the transfected cells using growth curve and soft agar assay respectively. Cell cycle analysis and apoptosis were determined by flow cytometry and cell death ELISA. The response to chemotherapeutic agents after transfecting anti-sense ITF was also examined. Anti-sense ITF transfectant (3A-5) had a significantly longer doubling time as compared to control cells which were transfected with empty vector (32.4 hr vs 26.9 hr, p < 0.05). In the soft agar assay, 3A-5 formed fewer colonies than control (3.5 colonies vs 23.5 colonies, p < 0.05). Although there was no significant difference in the cell cycle distribution between 3A-5 and control, anti-sense ITF resulted in marked increase in adriamycin-induced apoptosis. Our results demonstrated that blocking the expression of ITF inhibits growth of gastric cancer cells and enhances the response to chemotherapy.
Collapse
Affiliation(s)
- Michael W Y Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | | | | | | | | | | | | |
Collapse
|
21
|
Kato S, Matsukura N, Togashi A, Masuda G, Matsuda N, Yamada N, Naito Z, Matsuhisa T, Tajiri T. Sex differences in mucosal response to Helicobacter pylori infection in the stomach and variations in interleukin-8, COX-2 and trefoil factor family 1 gene expression. Aliment Pharmacol Ther 2004; 20 Suppl 1:17-24. [PMID: 15298601 DOI: 10.1111/j.1365-2036.2004.01985.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Gastric cancer incidence in men is almost double that in women. We investigated mucosal responses in the stomach against Helicobacter pylori (H. pylori) infections to elucidate the interindividual or sex-related differences, which may in turn be associated with gastric cancer incidence, mucosal changes of stomach as measured by the Sydney System, and interleukin-8, cyclooxygenase-2 and trefoil factor family 1 (TFF1) gene expression. METHODS An age-, sex-, H. pylori status- and disease-matched case-control study was performed in 574 H. pylori-positive and 225 H. pylori-negative patients selected from 4125 patients with a diagnosis of benign disease of the stomach. Levels of acute and chronic inflammations, atrophy and intestinal metaplasia scored according to the Sydney System were compared by stomach site and by sex. Two biopsy specimens (antral and corpus gastric mucosa) from patients with benign gastric diseases (142 patients; 72 men, 70 women) were analysed for interleukin-8, cyclooxygenase-2 and TFF1 mRNA expression as measured by real-time PCR. RESULTS Inflammation and activity scores in antrum with H. pylori infection were higher in men, but scores declined according to age. Atrophy and intestinal metaplasia scores in corpus with H. pylori infection appeared more severe in men than in women, especially in older patients. In women, atrophy score increased with increasing age, particularly in postmenopausal H. pylori-negative patients. Interleukin-8 mRNA induction was detected in both antrum and corpus mucosa in H. pylori infection, but sex differences were not found. Response of cyclooxygenase-2 mRNA expression against H. pylori infection in the mucosa was higher in men than women. In H. pylori-negative patients, TFF1 mRNA levels in women were significantly higher than in men, and TFF1 mRNA was significantly lower in positive than negative women. CONCLUSIONS Sex differences in mucosal responses to H. pylori infection in the stomach may be correlated with sex differences in the incidence of stomach cancer.
Collapse
Affiliation(s)
- S Kato
- Surgery for Organ Function and Biological Regulation, Nippon Medical School, Tama-Nagayama Hospital, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Leung WK, Yu J, Chan FKL, To KF, Chan MWY, Ebert MPA, Ng EKW, Chung SCS, Malfertheiner P, Sung JJY. Expression of trefoil peptides (TFF1, TFF2, and TFF3) in gastric carcinomas, intestinal metaplasia, and non-neoplastic gastric tissues. J Pathol 2002; 197:582-8. [PMID: 12210076 DOI: 10.1002/path.1147] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Trefoil factor family (TFF) domain peptides consist of three members that play a role in intestinal mucosal defence and repair, and in tumourigenesis. The role of the three TFF members in the gastric carcinogenesis cascade remains poorly defined. This study examined seven gastric cell lines, 50 gastric cancers and their adjacent non-cancer tissues, and tissues from 40 non-cancer patients, in order to elucidate the chronology of TFF expression in various stages of gastric carcinogenesis. TFF expression was determined by RT-PCR, immunohistochemistry, and western blot. Aberrant expression of TFF1, TFF2, and TFF3 was frequently detected in gastric cell lines. Specifically, TFF1 was detected in all non-cancer patients, but was detected in only 50% of gastric cancer and 66% of adjacent normal tissues. TFF2 expression was demonstrated in 87.5% of non-cancer patients, 34% of gastric carcinomas, and 58% of adjacent non-cancer tissues. There was a significant correlation between TFF1 and TFF2 expression in gastric cancer and adjacent non-cancer tissues (p<0.001). By contrast, TFF3 was detected in 25% of non-cancer patients and showed a predilection for areas with intestinal metaplasia (p=0.005). Sixty-two per cent of gastric cancers and 24% of neighbouring non-cancer tissues showed TFF3 expression. Immunoreactivity against TFF3 was demonstrated in goblet cells of intestinal metaplasia and within the cytoplasm and nuclei of tumour cells. Progressive loss of TFF1 and TFF2, together with the induction of TFF3, is likely to be involved in the early stage of the multi-step gastric carcinogenesis pathway.
Collapse
Affiliation(s)
- Wai K Leung
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Machado JC, Nogueira AM, Carneiro F, Reis CA, Sobrinho-Simões M. Gastric carcinoma exhibits distinct types of cell differentiation: an immunohistochemical study of trefoil peptides (TFF1 and TFF2) and mucins (MUC1, MUC2, MUC5AC, and MUC6). J Pathol 2000. [PMID: 10699992 DOI: 10.1002/(sici)1096-9896(200003)190:4%3c437::aid-path547%3e3.0.co;2-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The expression of trefoil peptides (TFF1 and TFF2) and mucins (MUC1, MUC2, MUC5AC, and MUC6) has previously been described in gastric polyps. In the present study, the expression profile of these trefoil peptides and mucins was characterized in 96 gastric carcinomas, in an attempt to further the understanding of the histogenesis and cell differentiation of gastric carcinoma. Taking together the co-expression of trefoil peptides and mucins, three phenotypes were defined: complete gastric, incomplete gastric, and non-gastric phenotype. Gastric differentiation (complete and incomplete) was observed in 30 out of 33 (90.9%) diffuse carcinomas and in 38 out of 53 (71.7%) intestinal carcinomas. Non-gastric differentiation was observed in only three (9.1%) diffuse carcinomas and in 15 (28.3%) intestinal carcinomas. The phenotypes observed in intestinal carcinomas were similar to those previously observed in adenomatous polyps, whereas most diffuse carcinomas mimicked the phenotype of hyperplastic polyps. The percentage of cases displaying a non-gastric phenotype was higher, though not significantly, in tumours that had invaded the gastric wall than in T1 tumours, regardless of histotype. It is concluded that gastric-type differentiation is retained in the majority of gastric carcinomas, being more prominent in diffuse than in intestinal carcinomas, and in early than in advanced carcinomas.
Collapse
Affiliation(s)
- J C Machado
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Roberto Frias s/n, 4200 Porto, Portugal.
| | | | | | | | | |
Collapse
|
24
|
Machado JC, Nogueira AM, Carneiro F, Reis CA, Sobrinho-Simões M. Gastric carcinoma exhibits distinct types of cell differentiation: an immunohistochemical study of trefoil peptides (TFF1 and TFF2) and mucins (MUC1, MUC2, MUC5AC, and MUC6). J Pathol 2000; 190:437-43. [PMID: 10699992 DOI: 10.1002/(sici)1096-9896(200003)190:4<437::aid-path547>3.0.co;2-1] [Citation(s) in RCA: 118] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The expression of trefoil peptides (TFF1 and TFF2) and mucins (MUC1, MUC2, MUC5AC, and MUC6) has previously been described in gastric polyps. In the present study, the expression profile of these trefoil peptides and mucins was characterized in 96 gastric carcinomas, in an attempt to further the understanding of the histogenesis and cell differentiation of gastric carcinoma. Taking together the co-expression of trefoil peptides and mucins, three phenotypes were defined: complete gastric, incomplete gastric, and non-gastric phenotype. Gastric differentiation (complete and incomplete) was observed in 30 out of 33 (90.9%) diffuse carcinomas and in 38 out of 53 (71.7%) intestinal carcinomas. Non-gastric differentiation was observed in only three (9.1%) diffuse carcinomas and in 15 (28.3%) intestinal carcinomas. The phenotypes observed in intestinal carcinomas were similar to those previously observed in adenomatous polyps, whereas most diffuse carcinomas mimicked the phenotype of hyperplastic polyps. The percentage of cases displaying a non-gastric phenotype was higher, though not significantly, in tumours that had invaded the gastric wall than in T1 tumours, regardless of histotype. It is concluded that gastric-type differentiation is retained in the majority of gastric carcinomas, being more prominent in diffuse than in intestinal carcinomas, and in early than in advanced carcinomas.
Collapse
Affiliation(s)
- J C Machado
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Roberto Frias s/n, 4200 Porto, Portugal.
| | | | | | | | | |
Collapse
|
25
|
Fujimoto J, Yasui W, Tahara H, Tahara E, Kudo Y, Yokozaki H, Tahara E. DNA hypermethylation at the pS2 promoter region is associated with early stage of stomach carcinogenesis. Cancer Lett 2000; 149:125-34. [PMID: 10737716 DOI: 10.1016/s0304-3835(99)00349-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
pS2, a member of the trefoil peptide family, has been suggested to be a gastric-specific tumor suppressor. We examined the expression of pS2 in gastric carcinomas, adenomas and non-neoplastic mucosa and analyzed the DNA methylation in the pS2 promoter. Reduced expression of pS2 was frequently associated with well-differentiated adenocarcinomas. The CpG sites within the promoter region of the pS2 gene were methylated in pS2-negative gastric carcinoma cell lines whereas it was not in pS2-positive cell line. The promoter methylation was detected in gastric carcinoma tissues and intestinal metaplasia with reduced pS2 expression whereas none of the carcinomas with preserved pS2 expression showed the promoter methylation. These findings suggest that reduced expression of pS2 due to the promoter methylation may participate in an early stage of stomach carcinogenesis, especially of well differentiated type.
Collapse
Affiliation(s)
- J Fujimoto
- First Department of Pathology, Hiroshima University School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
26
|
Calnan DP, Westley BR, May FE, Floyd DN, Marchbank T, Playford RJ. The trefoil peptide TFF1 inhibits the growth of the human gastric adenocarcinoma cell line AGS. J Pathol 1999; 188:312-7. [PMID: 10419601 DOI: 10.1002/(sici)1096-9896(199907)188:3<312::aid-path360>3.0.co;2-p] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
TFF1 is a 60-amino acid peptide produced in normal gastric mucosa which forms dimers spontaneously. Tumours of patients with gastric cancer usually have reduced TFF1 levels and disruption of the TFF1 gene causes animals to develop gastric adenomas and carcinomas. The effect of normal sequence human recombinant TFF1 and an analogue (Cys(58)-->Ser(58)), which is unable to dimerize, on the proliferation and morphology of the human gastric adenocarcinoma cell line AGS was therefore investigated. Proliferation, assessed by total cell number and [methyl-(3)H]thymidine incorporation, was reduced by dimeric TFF1 in a dose-dependent manner. Monomeric TFF1 also reduced proliferation but was less potent than the dimeric form. It is concluded that TFF1 may be an important controller of gastric cell proliferation, that dimerization of TFF1 is important in this effect, and that the reduced levels of TFF1 seen in gastric cancer may be of clinical relevance.
Collapse
Affiliation(s)
- D P Calnan
- University Division of Gastroenterology, Leicester General Hospital, Gwendolen Road, Leicester LE5 4PW, U.K
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Medullary thyroid carcinoma (MTC) is an uncommon tumour of calcitonin-secreting C-cells of the thyroid gland. This cancer represents an important potential model for the study of mechanisms of human epithelial cell transformation. Although recent studies have identified the gene involved in familial forms of MTC, little is known about the molecular pathogenesis of the sporadic variants of this tumour. The biological and prognostic significance of TFF1 expression, particularly in diverse human malignancies, suggests that the TFF1 protein could have a role in human neoplasia. Furthermore, in prostate cancer it has been demonstrated that TFF1 expression is closely associated with premalignant changes and neuroendocrine differentiation. In the present study, the expression of TFF1 was analysed in 18 human MTCs, comprising sporadic and familial tumours, C-cell hyperplasia, and one case of lymph gland metastasis. TFF1 expression was also examined in the cultures of a human MTC-derived tumour cell line (TT cell line). The results showed that ten sporadic tumours, three hereditary tumours (including C-cell hyperplasia), and one lymph gland metastasis displayed TFF1 immunoreactivity. Indirect fluorescence immunocytochemistry and Western blotting revealed that the TFF1 protein was strongly expressed in the TT cells. Northern analysis revealed that tumours and TT cells expressed the TFF1 transcript. Although the function of TFF1 protein in the carcinogenesis of MTC remains to be elucidated, its expression in the majority of cases of both sporadic and hereditary tumours, metastatic tumours, and in C-cell hyperplasia suggests that it may contribute to the pathogenesis of MTC.
Collapse
Affiliation(s)
- D G Wang
- Department of Medicine, School of Clinical Medicine, Queen's University of Belfast, U.K
| | | | | | | | | | | | | |
Collapse
|
28
|
Wu MS, Shun CT, Wang HP, Lee WJ, Wang TH, Lin JT. Loss of pS2 protein expression is an early event of intestinal-type gastric cancer. Jpn J Cancer Res 1998; 89:278-82. [PMID: 9600121 PMCID: PMC5921795 DOI: 10.1111/j.1349-7006.1998.tb00559.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
To investigate the prevalence of pS2 expression in gastric cancer with respect to tumor histopathology, intestinal metaplasia and Helicobacter pylori (H. pylori) infection, pathologic specimens of 91 patients with gastric cancer were immunostained for pS2. Such immunoreactivity was correlated with the status of H. pylori infection, tumor staging, histology, subtyping, and associated intestinal metaplasia. Positive pS2 staining was seen throughout all non-neoplastic epithelia, and in all 9 patients with the complete type of intestinal metaplasia. In contrast, 21 of 45 incomplete type of intestinal metaplasia had negative pS2 staining (P < 0.001), and 54 out of 91 tumors (59.3%) showed loss of pS2 expression in the cancer tissues proper. There was no correlation of pS2 expression with age, gender, depth of invasion, duodenal involvement, lymph node metastasis, venous invasion or H. pylori infection. Negative pS2 staining was significantly higher in the intestinal (74.5%) and Borrmann type I, II, III (64.2%) tumors than the diffuse (43.2%, P < 0.005) and Borrmann type IV (20%, P < 0.05) tumors. Our results indicate that loss of pS2 expression may occur as an early event in the malignant transformation process of intestinal-type tumors.
Collapse
Affiliation(s)
- M S Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei
| | | | | | | | | | | |
Collapse
|
29
|
Wang DG, Johnston CF, Liu WH, Sloan JM, Buchanan KD. Expression of a breast-cancer-associated protein (pS2) in human neuro-endocrine tumours. Int J Cancer 1997; 74:270-4. [PMID: 9221803 DOI: 10.1002/(sici)1097-0215(19970620)74:3<270::aid-ijc6>3.0.co;2-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
pS2 protein expression has been demonstrated in a range of malignant tissues in an oestrogen-independent pathway. Recently, it has been demonstrated that pS2, in prostate cancer, is closely associated with neuro-endocrine differentiation. In the present study, we have analyzed, by immunohistochemistry along with microwave antigen retrieval, the expression of pS2 protein in a retrospective series of 236 human primary neuro-endocrine tumours and attempted to correlate this with the clinicopathologic features of patients and the presence of oestrogen receptor (ER). pS2 immunoreactivity was detected in 42% of small-cell lung carcinomas, 36% of lung carcinoids, 33% of phaeochromocytomas, 38% of carotid-body tumours, 31% of pancreatic neuro-endocrine tumours, 60% of stomach carcinoids, 55% of ileal carcinoids, 23% of appendiceal carcinoids and 86% of rectal carcinoids respectively in more than 10% tumour cells. No pituitary tumours displayed pS2 immunoreactivity. pS2 transcript was also detected in lung carcinoid and carotid-body tumours by Northern-blot analysis. There was a statistically higher incidence of pS2 expression in carcinoid tumours of the ileum and rectum than in those of the appendix. No association was observed between pS2 expression and the occurrence of the carcinoid syndrome; nor was any correlation observed between the occurrence of pS2 immunoreactivity and that of ER. Our results suggest that the expression of the pS2 protein in a wide spectrum of neuro-endocrine tumours may be implicated in the pathogenesis and progression of some neuro-endocrine tumours in an oestrogen-independent pathway.
Collapse
Affiliation(s)
- D G Wang
- Metabolism and Endocrinology Division, School of Clinical Medicine, The Queen's University of Belfast, N. Ireland, UK.
| | | | | | | | | |
Collapse
|
30
|
|
31
|
Machado JC, Carneiro F, Ribeiro P, Blin N, Sobrinho-Simões M. pS2 protein expression in gastric carcinoma. An immunohistochemical and immunoradiometric study. Eur J Cancer 1996; 32A:1585-90. [PMID: 8911122 DOI: 10.1016/0959-8049(96)00116-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The aim of this study was to examine the prevalence of pS2 expression in gastric carcinoma and to determine its prognostic significance. We analysed pS2 protein expression in 50 gastric carcinomas and respective adjacent mucosas by immunohistochemistry and immunoradiometric assay (IRMA). pS2 was consistently expressed in superficial and foveolar epithelium of non-neoplastic mucosa and in 66.0% of the carcinomas. pS2 immunoreactivity was significantly higher in diffuse than in intestinal carcinomas, and in those cases with nodal metastases than in those without. No correlation was found between pS2 immunostaining and gender, age, staging, depth of wall penetration, venous invasion, ploidy and S-phase fraction. The mean levels of pS2 (IRMA) were significantly lower in gastric carcinomas than in non-neoplastic mucosas, and were not correlated with any of the aforementioned clinicopathological features. The survival of patients with pS2-positive tumours was not significantly different from that of patients with pS2-negative tumours. We conclude that pS2 expression, which can be used as a marker of gastric-type differentiation, is associated with gastric carcinoma of diffuse type. The lack of correlation between pS2 expression and most features of tumor aggressiveness and patients' survival precludes its use as a prognostic tool in gastric carcinoma.
Collapse
Affiliation(s)
- J C Machado
- Department of Pathology, IPATIMUP, Medical Faculty, University of Porto, Portugal
| | | | | | | | | |
Collapse
|
32
|
Theisinger B, Seitz G, Dooley S, Welter C. A second trefoil protein, ITF/hP1.B, is transcribed in human breast cancer. Breast Cancer Res Treat 1996; 38:145-51. [PMID: 8861832 DOI: 10.1007/bf01806668] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Trefoil proteins form a specific group of stable secreted polypeptides. They are expressed in a lot of human cancers and during inflammatory processes of the gastrointestinal tract. Recently a new human trefoil protein, ITF/hP1.B, was isolated. Until now no studies of the activity of this gene in human solid tumors exist. In our examination we show for the first time that this gene is transcribed in human breast cancer. In contrast to another trefoil protein, pS2, the expression of ITF/hP1.B is not under control of estrogen in the human breast cancer cell line MCF-7. We suggest that the gene activity of ITF/hP1.B in addition to pS2 expression may be an improved prognostic marker in human breast cancer.
Collapse
Affiliation(s)
- B Theisinger
- Institut fur Humangenetik, Universitat des Saarlandes, Germany
| | | | | | | |
Collapse
|
33
|
Abstract
A series of 201 bladder cancer biopsy specimens was analysed immunohistochemically for the expression of pS2 protein. Altogether, 61 per cent of the tumours were pS2-negative; in 16 per cent less than 1 per cent and in 23 per cent of cases more than 1 per cent of cells were pS2-positive. Normal transitional epithelium was negative for pS2. The fraction of positive cells was higher in poorly differentiated non-papillary tumours and in invasive tumours with pelvic lymph-node (P = 0.05) and distant metastasis (P = 0.10). pS2 expression was not related to sex, while patients aged 60-70 years had low fractions of pS2-positive cells (P = 0.03). DNA ploidy, S-phase fraction, mitotic index, morphometric nuclear features, and expression of c-erbB-2, p53, and epidermal growth factor receptor were independent of expression of pS2. Tumours expressing pS2 in over 10 per cent of cells had a lower survival probability (P = 0.0486). The results show that pS2 is expressed in 40 per cent of transitional cell bladder tumours, but that this marker has no clinical significance over established prognostic factors.
Collapse
Affiliation(s)
- P K Lipponen
- Department of Pathology, University of Kuopio, Finland
| | | |
Collapse
|