1
|
Ghanemi A, Yoshioka M, St-Amand J. Trefoil Factor Family Member 2: From a High-Fat-Induced Gene to a Potential Obesity Therapy Target. Metabolites 2021; 11:metabo11080536. [PMID: 34436477 PMCID: PMC8401738 DOI: 10.3390/metabo11080536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/11/2022] Open
Abstract
Obesity has its epidemiological patterns continuously increasing. With controlling both diet and exercise being the main approaches to manage the energy metabolism balance, a high-fat (HF) diet is of particular importance. Indeed, lipids have a low satiety potential but a high caloric density. Thus, focusing on pharmacologically targetable pathways remains an approach with promising therapeutic potential. Within this context, trefoil factor family member 2 (Tff2) has been characterized as specifically induced by HF diet rather than low-fat diet. TFF2 has also been linked to diverse neurological mechanisms and metabolic patterns suggesting its role in energy balance. The hypothesis is that TFF2 would be a HF diet-induced signal that regulates metabolism with a focus on lipids. Within this review, we put the spotlight on key findings highlighting this line of thought. Importantly, the hypothetical mechanisms pointed highlight TFF2 as an important contributor to obesity development via increasing lipids intestinal absorption and anabolism. Therefore, an outlook for future experimental activities and evaluation of the therapeutic potential of TFF2 inhibition is given. Indeed, its knockdown or downregulation would contribute to an antiobesity phenotype. We believe this work represents an addition to our understanding of the lipidic molecular implications in obesity, which will contribute to develop therapies aiming to manage the lipidic metabolic pathways including the absorption, storage and metabolism via targeting TFF2-related pathways. We briefly discuss important relevant concepts for both basic and clinical researchers.
Collapse
Affiliation(s)
- Abdelaziz Ghanemi
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Quebec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
| | - Mayumi Yoshioka
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Quebec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
| | - Jonny St-Amand
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Quebec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
- Correspondence:
| |
Collapse
|
2
|
CD147 receptor is essential for TFF3-mediated signaling regulating colorectal cancer progression. Signal Transduct Target Ther 2021; 6:268. [PMID: 34262017 PMCID: PMC8280106 DOI: 10.1038/s41392-021-00677-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
Major gaps in understanding the molecular mechanisms of colorectal cancer (CRC) progression and intestinal mucosal repair have hampered therapeutic development for gastrointestinal disorders. Trefoil factor 3 (TFF3) has been reported to be involved in CRC progression and intestinal mucosal repair; however, how TFF3 drives tumors to become more aggressive or metastatic and how TFF3 promotes intestinal mucosal repair are still poorly understood. Here, we found that the upregulated TFF3 in CRC predicted a worse overall survival rate. TFF3 deficiency impaired mucosal restitution and adenocarcinogenesis. CD147, a membrane protein, was identified as a binding partner for TFF3. Via binding to CD147, TFF3 enhanced CD147-CD44s interaction, resulting in signal transducer and activator of transcription 3 (STAT3) activation and prostaglandin G/H synthase 2 (PTGS2) expression, which were indispensable for TFF3-induced migration, proliferation, and invasion. PTGS2-derived PGE2 bound to prostaglandin E2 receptor EP4 subtype (PTGER4) and contributed to TFF3-stimulated CRC progression. Solution NMR studies of the TFF3-CD147 interaction revealed the key residues critical for TFF3 binding and the induction of PTGS2 expression. The ability of TFF3 to enhance mucosal restitution was weakened by a PTGS2 inhibitor. Blockade of TFF3-CD147 signaling using competitive inhibitory antibodies or a PTGS2 inhibitor reduced CRC lung metastasis in mice. Our findings bring strong evidence that CD147 is a novel receptor for TFF3 and PTGS2 signaling is critical for TFF3-induced mucosal restitution and CRC progression, which widens and deepens the understanding of the molecular function of trefoil factors.
Collapse
|
3
|
Braga Emidio N, Brierley SM, Schroeder CI, Muttenthaler M. Structure, Function, and Therapeutic Potential of the Trefoil Factor Family in the Gastrointestinal Tract. ACS Pharmacol Transl Sci 2020; 3:583-597. [PMID: 32832864 DOI: 10.1021/acsptsci.0c00023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Indexed: 12/20/2022]
Abstract
Trefoil factor family peptides (TFF1, TFF2, and TFF3) are key players in protecting, maintaining, and repairing the gastrointestinal tract. Accordingly, they have the therapeutic potential to treat and prevent a variety of gastrointestinal disorders associated with mucosal damage. TFF peptides share a conserved motif, including three disulfide bonds that stabilize a well-defined three-loop-structure reminiscent of a trefoil. Although multiple functions have been described for TFF peptides, their mechanisms at the molecular level remain poorly understood. This review presents the status quo of TFF research relating to gastrointestinal disorders. Putative TFF receptors and protein partners are described and critically evaluated. The therapeutic potential of these peptides in gastrointestinal disorders where altered mucosal biology plays a crucial role in the underlying etiology is discussed. Finally, areas of investigation that require further research are addressed. Thus, this review provides a comprehensive update on TFF literature as well as guidance toward future research to better understand this peptide family and its therapeutic potential for the treatment of gastrointestinal disorders.
Collapse
Affiliation(s)
- Nayara Braga Emidio
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medicial Research Insittitue (FHMRI), Flinders University, Bedford Park, South Australia 5042, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia.,Discipline of Medicine, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Christina I Schroeder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia.,National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Markus Muttenthaler
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
4
|
Jahan R, Shah A, Kisling SG, Macha MA, Thayer S, Batra SK, Kaur S. Odyssey of trefoil factors in cancer: Diagnostic and therapeutic implications. Biochim Biophys Acta Rev Cancer 2020; 1873:188362. [PMID: 32298747 DOI: 10.1016/j.bbcan.2020.188362] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 02/07/2023]
Abstract
Trefoil factors 1, 2, and 3 (TFFs) are a family of small secretory molecules involved in the protection and repair of the gastrointestinal tract (GI). TFFs maintain and restore epithelial structural integrity via transducing key signaling pathways for epithelial cell migration, proliferation, and invasion. In recent years, TFFs have emerged as key players in the pathogenesis of multiple diseases, especially cancer. Initially recognized as tumor suppressors, emerging evidence demonstrates their key role in tumor progression and metastasis, extending their actions beyond protection. However, to date, a comprehensive understanding of TFFs' mechanism of action in tumor initiation, progression and metastasis remains obscure. The present review discusses the structural, functional and mechanistic implications of all three TFF family members in tumor progression and metastasis. Also, we have garnered information from studies on their structure and expression status in different organs, along with lessons from their specific knockout in mouse models. In addition, we highlight the emerging potential of using TFFs as a biomarker to stratify tumors for better therapeutic intervention.
Collapse
Affiliation(s)
- Rahat Jahan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA
| | - Ashu Shah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA
| | - Sophia G Kisling
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA
| | - Muzafar A Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA; Department of Otolaryngology-Head & Neck Surgery, University of Nebraska Medical Center, NE, 68198, USA; Department of Biotechnology, Central University of Kashmir, Ganderbal, Jammu and Kashmir, India -191201
| | - Sarah Thayer
- Division of Surgical Oncology, Department of Surgery, University of Nebraska Medical Center, NE, 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, NE, 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, NE 68198, USA.
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA.
| |
Collapse
|
5
|
Zhu Y, Zhao S, Deng Y, Gordillo R, Ghaben AL, Shao M, Zhang F, Xu P, Li Y, Cao H, Zagnitko O, Scott DA, Gupta RK, Xing C, Zhang BB, Lin HV, Scherer PE. Hepatic GALE Regulates Whole-Body Glucose Homeostasis by Modulating Tff3 Expression. Diabetes 2017; 66:2789-2799. [PMID: 28877911 PMCID: PMC5652600 DOI: 10.2337/db17-0323] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 08/28/2017] [Indexed: 01/03/2023]
Abstract
Transcripts of key enzymes in the Leloir pathway of galactose metabolism in mouse livers are significantly increased after chronic high-fat/high-sucrose feeding. UDP-galactose-4-epimerase (GALE) is the last enzyme in this pathway that converts UDP-galactose to UDP-glucose and was previously identified as a downstream target of the endoplasmic reticulum (ER) stress effector spliced X-box binding protein 1, suggesting an interesting cross talk between galactose and glucose metabolism in the context of hepatic ER stress and whole-body metabolic fitness. However, its specific role in glucose metabolism is not established. Using an inducible and tissue-specific mouse model, we report that hepatic overexpression of Gale increases gluconeogenesis from pyruvate and impairs glucose tolerance. Conversely, genetic reduction of Gale in liver improves glucose tolerance. Transcriptional profiling identifies trefoil factor 3 (Tff3) as one of the downstream targets of GALE. Restoration of Tff3 expression corrects glucose intolerance in Gale-overexpressing mice. These studies reveal a new link between hepatic GALE activity and whole-body glucose homeostasis via regulation of hepatic Tff3 expression.
Collapse
Affiliation(s)
- Yi Zhu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN
- Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX
| | - Shangang Zhao
- Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX
| | - Yingfeng Deng
- Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX
| | - Ruth Gordillo
- Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX
| | | | - Mengle Shao
- Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX
| | - Fang Zhang
- Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ping Xu
- Lilly China Research and Development Center, Shanghai, China
| | - Yang Li
- Lilly China Research and Development Center, Shanghai, China
| | - Huachuan Cao
- Lilly China Research and Development Center, Shanghai, China
| | - Olga Zagnitko
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - David A Scott
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Rana K Gupta
- Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX
| | - Chao Xing
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX
| | - Bei B Zhang
- Lilly China Research and Development Center, Shanghai, China
| | - Hua V Lin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN
| | - Philipp E Scherer
- Touchstone Diabetes Center, UT Southwestern Medical Center, Dallas, TX
| |
Collapse
|
6
|
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
|
7
|
Abstract
Necrotising enterocolitis (NEC) is a serious infection of the bowel that predominantly affects preterm infants and is a leading cause for mortality and morbidity in preterm infants. It involves a spectrum of pathology including widespread inflammation of the intestinal mucosa, invasion of the immature gut by enteric gas forming bacteria, dissection of the gut wall and portal veins by this gas, often culminating in ischemic necrosis of the intestine. This article provides an overview of the incidence, etio-pathological risk factors, preventive strategies and medical management of NEC.
Collapse
|
8
|
Ge H, Gardner J, Wu X, Rulifson I, Wang J, Xiong Y, Ye J, Belouski E, Cao P, Tang J, Lee KJ, Coberly S, Wu X, Gupte J, Miao L, Yang L, Nguyen N, Shan B, Yeh WC, Véniant MM, Li Y, Baribault H. Trefoil Factor 3 (TFF3) Is Regulated by Food Intake, Improves Glucose Tolerance and Induces Mucinous Metaplasia. PLoS One 2015; 10:e0126924. [PMID: 26083576 PMCID: PMC4471263 DOI: 10.1371/journal.pone.0126924] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 04/09/2015] [Indexed: 02/06/2023] Open
Abstract
Trefoil factor 3 (TFF3), also called intestinal trefoil factor or Itf, is a 59 amino acid peptide found as a homodimer predominantly along the gastrointestinal tract and in serum. TFF3 expression is elevated during gastrointestinal adenoma progression and has been shown to promote mucosal wound healing. Here we show that in contrast to other trefoil factor family members, TFF1 and TFF2, TFF3 is highly expressed in mouse duodenum, jejunum and ileum and that its expression is regulated by food intake. Overexpression of TFF3 using a recombinant adeno-associated virus (AAV) vector, or daily administration of recombinant TFF3 protein in vivo improved glucose tolerance in a diet-induced obesity mouse model. Body weight, fasting insulin, triglyceride, cholesterol and leptin levels were not affected by TFF3 treatment. Induction of mucinous metaplasia was observed in mice with AAV-mediated TFF3 overexpression, however, no such adverse histological effect was seen after the administration of recombinant TFF3 protein. Altogether these results suggest that the therapeutic potential of targeting TFF3 to treat T2D may be limited.
Collapse
Affiliation(s)
- Hongfei Ge
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
| | - Jonitha Gardner
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
| | - Xiaosu Wu
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
| | - Ingrid Rulifson
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
| | - Jinghong Wang
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
| | - Yumei Xiong
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
| | - Jingjing Ye
- Amgen, Protein Technologies, South San Francisco, California, United States of America
| | - Edward Belouski
- Amgen, Protein Technologies, South San Francisco, California, United States of America
| | - Ping Cao
- Amgen, Protein Technologies, South San Francisco, California, United States of America
| | - Jie Tang
- Amgen, Protein Technologies, South San Francisco, California, United States of America
| | - Ki Jeong Lee
- Amgen, Lead Discovery, Thousand Oaks, California, United States of America
| | - Suzanne Coberly
- Amgen, Pathology, South San Francisco, California, United States of America
| | - Xinle Wu
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
| | - Jamila Gupte
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
| | - Lynn Miao
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
| | - Li Yang
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
| | - Natalie Nguyen
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
| | - Bei Shan
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
| | - Wen-Chen Yeh
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
| | - Murielle M. Véniant
- Amgen, Metabolic Disorders, Thousand Oaks, California, United States of America
| | - Yang Li
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
| | - Helene Baribault
- Amgen, Metabolic Disorders, South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
9
|
Trefoil factor 3 as an endocrine neuroprotective factor from the liver in experimental cerebral ischemia/reperfusion injury. PLoS One 2013; 8:e77732. [PMID: 24204940 PMCID: PMC3799633 DOI: 10.1371/journal.pone.0077732] [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: 05/02/2013] [Accepted: 09/08/2013] [Indexed: 01/30/2023] Open
Abstract
Cerebral ischemia, while causing neuronal injury, can activate innate neuroprotective mechanisms, minimizing neuronal death. In this report, we demonstrate that experimental cerebral ischemia/reperfusion injury in the mouse causes upregulation of the secretory protein trefoil factor 3 (TFF3) in the hepatocyte in association with an increase in serum TFF3. Partial hepatectomy (~60% liver resection) immediately following cerebral injury significantly lowered the serum level of TFF3, suggesting a contribution of the liver to the elevation of serum TFF3. Compared to wild-type mice, TFF3-/- mice exhibited a significantly higher activity of caspase 3 and level of cell death in the ischemic cerebral lesion, a larger fraction of cerebral infarcts, and a smaller fraction of the injured cerebral hemisphere, accompanied by severer forelimb motor deficits. Intravenous administration of recombinant TFF3 reversed changes in cerebral injury and forelimb motor function due to TFF3 deficiency. These observations suggest an endocrine neuroprotective mechanism involving TFF3 from the liver in experimental cerebral ischemia/reperfusion injury.
Collapse
|
10
|
Yong Z, Lin W, Yong S, Guang-ping L, Dan W, Shang-jun L, Wei W, Xi P. Kinetic characterization of an intestinal trefoil factor receptor. PLoS One 2013; 8:e74669. [PMID: 24086361 PMCID: PMC3781110 DOI: 10.1371/journal.pone.0074669] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 08/03/2013] [Indexed: 11/18/2022] Open
Abstract
Objective To determine whether intestinal epithelial cells have a receptor for intestinal trefoil factor and characterize receptor-ligand binding kinetics. Methods Radioligand binding assays were performed to characterize the binding kinetics between [125I]-labeled ITF and IEC-6, HT-29, Caco2 and HaCaT cells. The Kd, Bmax and other kinetic variables describing the interaction between ITF and its potential receptors were determined. Results Radioligand binding assays performed at 4°C showed that the Kd value for the association between [125I]-ITF and IEC-6, HT-29, and Caco2 cells were 1.99±0.12×10−9 M, 3.89±0.42×10−9 M, and 2.04±0.17×10−9 M, respectively. Bmax values were 1.17±0.04×1011, 3.97±0.29×1011, and 2.03±0.08×1011 sites/cell, respectively. The Ki values for the interaction between IEC-6, HT-29, and Caco2 cells and non-labeled ITF were 20.98±0.57 nM, 36.87±3.35 nM, and 21.38±0.93 nM, respectively, and the IC50 values were 25.21±0.39 nM, 40.68±0.27 nM, and 23.61±0.25 nM, respectively. Radioligand binding kinetic results showed the association rate constants (k+1) for IEC-6, HT-29, and Caco2 cells were 0.22±0.04 min−1, 0.29±0.04 min−1, and 0.26±0.05 min−1, respectively, and the dissociation rate constants (k-1) were 0.06±0.02 min−1, 0.03±0.01 min−1, and 0.04±0.01 min−1, respectively. For the HaCaT cells, the Kd was 4.86±0.28×10−8 M and Bmax was 5.81±0.15×108 sites/cell, the very low specific binding between [125I]-ITF and these cells made it impossible to calculate binding kinetic parameters. Conclusions An ITF-specific receptor appears to be present on the three types of intestinal epithelial cells (IEC-6, HT-29, and Caco-2), and there may be no ITF receptor on epidermal cells.
Collapse
Affiliation(s)
- Zhang Yong
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burns of PLA, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - Wang Lin
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burns of PLA, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - Sun Yong
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burns of PLA, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - Liang Guang-ping
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burns of PLA, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - Wu Dan
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burns of PLA, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - Lv Shang-jun
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burns of PLA, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - Wu Wei
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burns of PLA, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | - Peng Xi
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burns of PLA, Southwest Hospital, Third Military Medical University, Chongqing, PR China
- * E-mail:
| |
Collapse
|
11
|
Sun Y, Zhu Y, Wang L, Mao X, Peng X, Peng Y. Recombinant adenovirus-mediated intestinal trefoil factor gene therapy for burn-induced intestinal mucosal injury. PLoS One 2013; 8:e62429. [PMID: 23638081 PMCID: PMC3634752 DOI: 10.1371/journal.pone.0062429] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 03/21/2013] [Indexed: 02/07/2023] Open
Abstract
Intestinal trefoil factor (ITF) is a small polypeptide with potential medical values whose main pharmacological effects are to alleviate gastrointestinal mucosal injury caused by various injury factors and promote the repair of damaged mucosa. However, its low yield limits its application. The purpose of our study was to construct a recombinant adenoviral vector containing the hITF gene and observe the therapeutic effect of burn-induced intestinal mucosal injury using in vitro and in vivo analysis. First, a recombinant shuttle plasmid was constructed by ligating a pAdTrack-CMV vector with a full-length hITF gene containing a signal peptide and the mature peptide, followed by the recombinant Ad-hITF adenovirus vector after linearization and homologous recombination with the backbone plasmid in the competent BJ5183 strain. Second, the hITF expression level was detected using reverse transcription polymerase chain reaction and western blotting after Ad-hITF infection of colon cancer HT-29 cells. The recombinant adenovirus significantly promoted cell migration in an in vitro wounding model. Finally, we confirmed that the recombinant adenovirus could significantly expedite the healing of intestinal mucosal injury after establishing a mouse model in which severe burns were stimulated and the recombinant adenovirus was delivered by intragastric injection. In summary, we constructed a recombinant adenoviral vector containing the hITF gene and confirmed its role in promoting repair of the intestinal mucosa. Our study provides a novel way to treat burn-induced intestinal mucosal injury.
Collapse
Affiliation(s)
- Yong Sun
- Department of Burn Surgery, Plastic Surgery Center, No. 97 Hospital of PLA, Xuzhou, China
- * E-mail: (YS); (YZP)
| | - Yun Zhu
- Department of Gastroenterology, No. 97 Hospital of PLA, Xuzhou, China
| | - Liangxi Wang
- Department of Burn Surgery, Plastic Surgery Center, No. 97 Hospital of PLA, Xuzhou, China
| | - Xuefei Mao
- Department of Burn Surgery, Plastic Surgery Center, No. 97 Hospital of PLA, Xuzhou, China
| | - Xi Peng
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Yizhi Peng
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, The Third Military Medical University, Chongqing, China
- * E-mail: (YS); (YZP)
| |
Collapse
|
12
|
Shi HS, Yin X, Song L, Guo QJ, Luo XH. Neuropeptide Trefoil factor 3 improves learning and retention of novel object recognition memory in mice. Behav Brain Res 2012; 227:265-9. [DOI: 10.1016/j.bbr.2011.10.051] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Accepted: 10/31/2011] [Indexed: 12/17/2022]
|
13
|
Abstract
Necrotizing enterocolitis (NEC) is a common gastrointestinal inflammatory necrosis affecting almost exclusively premature infants usually after oral nutrition has been started, for example, 10 day plus postpartum. Although the pathogenesis is incompletely understood, major risk factors include prematurity and incomplete bacterial colonization. Evidence has been shown that the premature infant because of rapid passage through the birth canal or because of delivery by cesarean section has an inadequate initial ingestion of maternal colonic and vaginal flora and therefore, an inadequate initial colonization with less diversity of bacteria phylla and fewer species of bacteria in the microbiota. As a result, they are more susceptible to environmental pathogens. In addition, prematures have immature intestinal defenses (glycocalyx, tight junctions, innate immune response, etc.) resulting in excessive inflammation in response to luminal stimuli. Recently, we reported that genes mediating the innate inflammatory immune response are developmentally expressed with an increase in toll-like receptors, signaling molecules and transgenic factors and decreased negative regulators of inflammation, which undoubtedly contribute to an excessive inflammatory response. Several clinical studies have suggested that the use of probiotics and ingestion of expressed maternal breast milk containing probiotics can help to stabilize colonization and to reduce the incidence and severity of NEC when given to premature infants at risk. Meta-analyses of multiple small studies strongly suggest a protective effect in the use of probiotics. A multicenter study in Taiwan suggests that Bifidobacteria infantis and Lactobacillus acidophilus in combination may prevent NEC. These meta-analyses suggest that these probiotics should be used in routine care of premature infants. Other clinicians, however, suggest caution, holding out for a single protocol multicenter trial before routine use can be suggested.
Collapse
|
14
|
Claud EC. Probiotics and neonatal necrotizing enterocolitis. Anaerobe 2011; 17:180-5. [DOI: 10.1016/j.anaerobe.2011.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 01/28/2011] [Accepted: 02/03/2011] [Indexed: 01/15/2023]
|
15
|
Shi H, Ren J, Xu H, Pan J, Hao X, Barlow LL, Dong W. Upregulated expression of hITF in Crohn's disease and screening of hITF Interactant by a yeast two-hybrid system. Dig Dis Sci 2010; 55:2929-2939. [PMID: 20499178 DOI: 10.1007/s10620-010-1227-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Accepted: 03/25/2010] [Indexed: 12/16/2022]
Abstract
AIMS To study the expression of human intestinal trefoil factor (hITF) mRNA in Crohn's disease and to screen the cellular proteins that can interact with the hITF protein by a yeast two-hybrid system in order to explore the mechanism of hITF in protecting intestinal mucosa from injury. METHODS Seventy-eight patients underwent double-balloon enteroscopy (DBE). Expression of hITF mRNA was detected by quantitative real-time polymerase chain reaction analysis (qRT-PCR). The hITF gene was amplified by PCR and cloned into vector pDEST32. The yeast cells cotransformed with pDEST32-hITF and the human jejunal cDNA library were plated in a selective SC-Leu-Trp-His-Ura medium. The subsequent screen was performed with χ-gal detection, and true-positive clones were sequenced and analyzed with bioinformatics. Co-immunoprecipitation (Co-IP) was performed to confirm the binding of putative proteins to the hITF protein. RESULTS Thirty-nine patients were diagnosed with Crohn's disease. We found that the expression of hITF mRNA is significantly increased in Crohn's disease compared to normal controls. A total of ten colonies were selected and sequenced. Among these, six colonies were Homo sapiens zinc finger protein 193 (ZNF193), three colonies were Homo sapiens Aldo-keto reductase family 1C 1 (AKR1C1), and one colony was of an unknown gene. A reverse two-hybrid experiment and Co-IP indicated that ZNF193 and AKR1C1 might interact with hITF. CONCLUSIONS The expression of hITF mRNA is increased in Crohn's disease. ZNF193 and AKR1C1 are proteins that can interact with the hITF protein by a yeast two-hybrid system and Co-IP, hITF may contribute to the mucosal repair through this interaction.
Collapse
Affiliation(s)
- Huaxiu Shi
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | | | | | | | | | | | | |
Collapse
|
16
|
Dubeykovskaya Z, Dubeykovskiy A, Solal-Cohen J, Wang TC. Secreted trefoil factor 2 activates the CXCR4 receptor in epithelial and lymphocytic cancer cell lines. J Biol Chem 2008; 284:3650-62. [PMID: 19064997 DOI: 10.1074/jbc.m804935200] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The secreted trefoil factor family 2 (TFF2) protein contributes to the protection of the gastrointestinal mucosa from injury by strengthening and stabilizing mucin gels, stimulating epithelial restitution, and restraining the associated inflammation. Although trefoil factors have been shown to activate signaling pathways, no cell surface receptor has been directly linked to trefoil peptide signaling. Here we demonstrate the ability of TFF2 peptide to activate signaling via the CXCR4 chemokine receptor in cancer cell lines. We found that both mouse and human TFF2 proteins (at approximately 0.5 microm) activate Ca2+ signaling in lymphoblastic Jurkat cells that could be abrogated by receptor desensitization (with SDF-1alpha) or pretreatment with the specific antagonist AMD3100 or an anti-CXCR4 antibody. TFF2 pretreatment of Jurkat cells decreased Ca2+ rise and chemotactic response to SDF-1alpha. In addition, the CXCR4-negative gastric epithelial cell line AGS became highly responsive to TFF2 treatment upon expression of the CXCR4 receptor. TFF2-induced activation of mitogen-activated protein kinases in gastric and pancreatic cancer cells, KATO III and AsPC-1, respectively, was also dependent on the presence of the CXCR4 receptor. Finally we demonstrate a distinct proliferative effect of TFF2 protein on an AGS gastric cancer cell line that expresses CXCR4. Overall these data identify CXCR4 as a bona fide signaling receptor for TFF2 and suggest a mechanism through which TFF2 may modulate immune and tumorigenic responses in vivo.
Collapse
Affiliation(s)
- Zinaida Dubeykovskaya
- Department of Medicine, Columbia University Medical Center, New York, New York 10032, USA
| | | | | | | |
Collapse
|
17
|
Abstract
The intestinal microflora has a significant role in intestinal health and gut function. The neonatal population is unique in that intestinal colonization is not established and is known to be influenced by delivery method, feeding, gestational age, and medical interventions. The preterm infant is particularly sensitive to colonization patterns as inherent intestinal defense mechanisms are immature and immature intestinal epithelial cells are known to have exaggerated inflammatory responses to both commensal and pathogenic bacteria. These responses contribute to the development of neonatal necrotizing enterocolitis in this patient population. As certain bacteria are known to influence intestinal maturation and down-regulate intestinal inflammation, it has been suggested that influencing the intestinal flora of preterm infants may be beneficial. Clinical studies indicate that probiotic therapy may decrease the incidence of necrotizing enterocolitis and studies are ongoing to elucidate the mechanism of action of different probiotic organisms. Although concerns remain and further study is necessary, probiotics are a plausible means of optimizing intestinal colonization and influencing outcome of these vulnerable infants.
Collapse
|
18
|
Feng ZM, Fang DC, Chen WS, Wang RQ. Rodent IRR-219 (IgGFcgammaBP) and rTFF3, expressed mainly in the intestinal mucosa, depleted during dextran sulfate sodium-induced colitis. Dig Dis Sci 2007; 52:2104-12. [PMID: 17436098 DOI: 10.1007/s10620-006-9711-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Accepted: 11/27/2006] [Indexed: 12/31/2022]
Abstract
IgGFcgammaBP and TFF3 are related with adaptation during injury, mucosal defense, and epithelial healing. In this work, we produced the polyclonal antibodies for rat IgGFcgammaBP or TFF3 and assessed their tissue distributions in adult and prenatal rats, rTFF3 molecular patterns under reduced and nonreduced condition, involvement of IgGFcgammaBP, and TFF3 in dextran sulfate sodium (DSS)-induced colitis. Polyclonal antibodies of rat IgGFcgammaBP or TFF3 were produced with their synthetic polypeptide. Rat TFF3 was detected in the scraped intestinal mucosa by SDS/PAGE and Western blotting. Immunohistochemical stainings of rat IgGFcgammaBP or TFF3 were performed in different tissues, mainly in mucin-producing tissues, of adult rat and prenatal rat intestine. Rat IgGFcgammaBP and TFF3 were immunohistochemically detected in the distal colon of rat colitis model induced with 7% DSS. IgGFcgammaBP and TFF3 were mainly expressed in the intestinal mucosa with different distribution patterns. The scattered staining was also found in the epithelium of bile duct. There was strong expression of IgGFcgammaBP and TFF3 in rat embryonic intestine. There were two kinds of rTFF3 complexes existed with different molecular weights, 250 and 55 kDa, under nonreduced conditions, but shifted to 6 kDa under reduced conditions. In the DSS-induced colitis model, IgGFcgammaBP and TFF3 were significantly decreased in the distal colon mucosa at the onset and active phases comparing with the normal control, partially recovered at the regenerative phase. Based on these findings,IgGFcgammaBP and TFF3 were widely expressed in the intestinal mucosa, depleted during DSS-induced colitis. Rat TFF3 existed mainly in two complexes with 250 and 55 kDa molecular weights. The present findings indicate they are two important goblet cell-derived components possibly related to the pathogenesis of DSS-induced colitis, a rat model of ulcerative colitis.
Collapse
Affiliation(s)
- Zao-Ming Feng
- Department of Gastroenterology, Southwestern Hospital, Third Military Medical University, Chongqing 400038, People's Republic of China
| | | | | | | |
Collapse
|
19
|
Ren JL, Luo JY, Lu YP, Wang L, Shi HX. Molecular forms of trefoil factor 1 in normal gastric mucosa and its expression in normal and abnormal gastric tissues. World J Gastroenterol 2006; 12:7361-4. [PMID: 17143957 PMCID: PMC4087499 DOI: 10.3748/wjg.v12.i45.7361] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the molecular forms of trefoil factor 1 (TFF1) in normal gastric mucosa and its expression in normal and abnormal gastric tissues (gastric carcinoma, atypical hyperplasia and intestinalized gastric mucosa) and the role of TFF1 in the carcinogenesis and progression of gastric carcinoma and its molecular biological mechanism underlying gastric mucosa protection.
METHODS: The molecular forms of TFF1 in normal gastric mucosa were observed by Western blot. The expression of TFF1 in normal and abnormal gastric tissues (gastric carcinoma, atypical hyperplasia and intestinalized gastric mucosa) was also assayed by immunohistochemical method. The average positive AO was estimated by Motic Images Advanced 3.0 software.
RESULTS: Three patterns of TFF1 were found in normal gastric mucosa: monomer, dimmer, and TFF1 compound whose molecular weight is about 21 kDa. The concentration of TFF1 compound was the highest among these three patterns. TFF1 was expressed mainly in epithelial cytoplasm of the mucosa in gastric body and antrum, especially around the nuclei. The closer the TFF1 to the lumen, the higher the expression of TFF1. The expression of TFF1 in peripheral tissue of gastric carcinoma (0.51 ± 0.07) was higher than that in normal gastric mucosa (0.44 ± 0.06, P < 0.001). The expression of TFF1 in gastric adenocarcinoma was positively related to the differentiation of adenocarcinoma. The lower the differentiation of adenocarcinoma was, the weaker the expression of TFF1. No TFF1 was expressed in poorly-differentiated adenocarcinoma. The expression of TFF1 in moderately-well differentiated adenocarcinoma (0.45 ± 0.07) was a little lower than that in normal mucosa (P > 0.05). The expression of TFF1 in gastric mucosa with atypical hyperplasia (0.57 ± 0.03) was significantly higher than that in normal gastric mucosa (P < 0.001). No TFF1 was expressed in intestinalized gastric mucosa. There was no statistically significant difference between the expressions of TFF1 in gastric mucosa around the intestinalized tissue (0.45 ± 0.07) and normal gastric mucosa (P > 0.05).
CONCLUSION: TFF1 is expressed mainly in epithelial cytoplasm of the mucosa in gastric body and antrum. Its main pattern is TFF1 compound, which may have a greater biological activity than monomer and dimer. The expression of TFF1 in peripheral mucosa of gastric ulcer is higher than that in mucosa 5 cm beyond the ulcer, indicating that TFF1 plays an important part in protection and restitution of gastric mucosa. The expression of TFF1 is increased in peripheral tissues of gastric carcinoma and gastric mucosa with atypical hyperplasia, but is decreased in cancer tissues, implying that TFF1 may be related to suppression and differentiation of carcinoma. The weaker expression of TFF1 in poorly-differentiated carcinoma may be related to the destruction of glands and cells in cancer tissues and the decrease in secretion of TFF1.
Collapse
Affiliation(s)
- Jian-Lin Ren
- Department of Gastroenterology, The Second Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | | | | | | | | |
Collapse
|
20
|
Krop I, Parker MT, Bloushtain-Qimron N, Porter D, Gelman R, Sasaki H, Maurer M, Terry MB, Parsons R, Polyak K. HIN-1, an inhibitor of cell growth, invasion, and AKT activation. Cancer Res 2005; 65:9659-69. [PMID: 16266985 DOI: 10.1158/0008-5472.can-05-1663] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The HIN-1 gene encoding a small, secreted protein is silenced due to methylation in a substantial fraction of breast, prostate, lung, and pancreatic carcinomas, suggesting a potential tumor suppressor function. The receptor of HIN-1 is unknown, but ligand-binding studies indicate the presence of high-affinity cell surface HIN-1 binding on epithelial cells. Here, we report that HIN-1 is a potent inhibitor of anchorage-dependent and anchorage-independent cell growth, cell migration, and invasion. Expression of HIN-1 in synchronized cells inhibits cell cycle reentry and the phosphorylation of the retinoblastoma protein (Rb), whereas in exponentially growing cells, HIN-1 induces apoptosis without apparent cell cycle arrest and effect on Rb phosphorylation. Investigation of multiple signaling pathways revealed that mitogen-induced phosphorylation and activation of AKT are inhibited in HIN-1-expressing cells. In addition, expression of constitutively activate AKT abrogates HIN-1-mediated growth arrest. Taken together, these studies provide further evidence that HIN-1 possesses tumor suppressor functions, and that these activities may be mediated through the AKT signaling pathway.
Collapse
Affiliation(s)
- Ian Krop
- Department of Medical Oncology and Biostatistics, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
|
22
|
Zhu YQ, Tan XD. TFF3 modulates NF-{kappa}B and a novel negative regulatory molecule of NF-{kappa}B in intestinal epithelial cells via a mechanism distinct from TNF-{alpha}. Am J Physiol Cell Physiol 2005; 289:C1085-93. [PMID: 16014704 PMCID: PMC2527239 DOI: 10.1152/ajpcell.00185.2005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Trefoil factor 3 (intestinal trefoil factor) is a cytoprotective factor in the gut. Herein we compared the effect of trefoil factor 3 with tumor necrosis factor-alpha on 1) activation of NF-kappaB in intestinal epithelial cells; 2) expression of Twist protein (a molecule essential for downregulation of nuclear factor-kappaB activity in vivo); and 3) production of interleukin-8. We showed that Twist protein is constitutively expressed in intestinal epithelial cells. Tumor necrosis factor-alpha induced persistent degradation of Twist protein in intestinal epithelial cells via a signaling pathway linked to proteasome, which was associated with prolonged activation of NF-kappaB. In contrast to tumor necrosis factor, trefoil factor 3 triggered transient activation of NF-kappaB and prolonged upregulation of Twist protein in intestinal epithelial cells via an ERK kinase-mediated pathway. Unlike tumor necrosis factor-alpha, transient activation of NF-kappaB by trefoil factor 3 is not associated with induction of IL-8 in cells. To examine the role of Twist protein in intestinal epithelial cells, we silenced the Twist expression by siRNA. Our data showed that trefoil factor 3 induced interleukin-8 production after silencing Twist in intestinal epithelial cells. Together, these observations indicated that 1) trefoil factor 3 triggers a diverse signal from tumor necrosis factor-alpha on the activation of NF-kappaB and its associated molecules in intestinal epithelial cells; and 2) trefoil factor 3-induced Twist protein plays an important role in the modulation of inflammatory cytokine production in intestinal epithelial cells.
Collapse
Affiliation(s)
- Ya-Qin Zhu
- Molecular and Cellular Pathobiology Program, Children’s Memorial Research Center, Chicago, IL 60614
- Departments of Pathology and Pediatrics, Children’s Memorial Hospital, Feinberg School of Medicine, Northwestern University, Chicago, IL 60614
- Ya-Qin Zhu, M.D., functions as the co-corresponding author and could be contacted at
for the correspondence and requesting reprints
| | - Xiao-Di Tan
- Molecular and Cellular Pathobiology Program, Children’s Memorial Research Center, Chicago, IL 60614
- Departments of Pathology and Pediatrics, Children’s Memorial Hospital, Feinberg School of Medicine, Northwestern University, Chicago, IL 60614
- Address all correspondence to Xiao-Di Tan, M.D. Molecular and Cellular Pathobiology Program, Children’s Memorial Research Center, Children’s Memorial Hospital, 2300 Children’s Plaza, Box 217, Chicago, IL 60614, Tel: (773) 755-6380, Fax: (773) 755-6581, e-mail:
| |
Collapse
|
23
|
Poulsen SS, Kissow H, Hare K, Hartmann B, Thim L. Luminal and parenteral TFF2 and TFF3 dimer and monomer in two models of experimental colitis in the rat. ACTA ACUST UNITED AC 2005; 126:163-71. [PMID: 15664663 DOI: 10.1016/j.regpep.2004.09.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2004] [Revised: 08/26/2004] [Accepted: 09/19/2004] [Indexed: 01/11/2023]
Abstract
BACKGROUND Peptides of the trefoil factor family (TFF1, TFF2 and TFF3) are cosecreted with mucus from mucus-producing cells in most organ systems and are believed to interact with mucus to form high-viscosity stable gel complexes. In the gastrointestinal tract, they sustain the mucosal barrier, and both injected and orally administered TFF peptide have protective and healing functions in the gastric mucosa. AIM To investigate the possible treatment effect of luminally and parenterally administered TFF peptides in experimental colitis in rats. METHODS Colitis was induced by administration of 5% dextran sodium sulphate in the drinking water or by one intraperitoneal injection of mitomycin C, 3.75 mg/kg. TFF peptides were administered as subcutaneous injections or directly into the lumen via a catheter placed in the proximal colon. Treatments were saline, TFF2, TFF3 monomer or TFF3 dimer 5 mg/kg twice per day throughout the study [dextran sulphate sodium (DSS)] or from day 4 to 7 (mitomycin C). Colitis severity was scored in a stereomicroscope and histologically. RESULTS Luminal treatment with TFF3 in its dimeric form significantly improved the colitis score in both colitis models, whereas TFF2 had positive effect only in DSS-induced colitis. The TFF3 monomer was without any effects in both models. Treatment effect was most pronounced in the middle part of the colon, closest to the tip of the catheter. Injected TFF peptides, especially the TFF3 monomer, aggravated the colitis score in both colitis models. CONCLUSIONS Intracolonic administration of TFF3 dimer and TFF2 improves experimentally induced colitis in rats. The TFF3 monomer has no effect. Parenteral administration of TFF peptides aggravates the colitis especially the TFF3 monomer.
Collapse
|
24
|
N/A. N/A. Shijie Huaren Xiaohua Zazhi 2004; 12:2186-2188. [DOI: 10.11569/wcjd.v12.i9.2186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
25
|
Emami S, Rodrigues S, Rodrigue CM, Le Floch N, Rivat C, Attoub S, Bruyneel E, Gespach C. Trefoil factor family (TFF) peptides and cancer progression. Peptides 2004; 25:885-98. [PMID: 15177885 DOI: 10.1016/j.peptides.2003.10.019] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2003] [Accepted: 10/27/2003] [Indexed: 12/15/2022]
Abstract
TFF peptides are involved in mucosal maintenance and repair through motogenic and antiapoptotic activities. These peptides are overexpressed during inflammatory processes and cancer progression. They also function as scatter factors, proinvasive and angiogenic agents. Such a divergence is related to the pathophysiological state of tissues submitted to persistent aggressive situations during digestive processes in the normal gastrointestinal tract, inflammatory and neoplastic diseases. In agreement with this model, TFF peptides are connected with multiple oncogenic pathways. As a consequence, the TFF signaling pathways may serve as potential targets in the control of chronic inflammation and progression of human solid tumors.
Collapse
Affiliation(s)
- Shahin Emami
- INSERM U482, Signal Transduction and Cellular Functions in Diabetes and Digestive Cancers, Hôpital Saint-Antoine, 75571 Paris Cedex 12, France.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Poulsen SS, Thulesen J, Hartmann B, Kissow HL, Nexø E, Thim L. Injected TFF1 and TFF3 bind to TFF2-immunoreactive cells in the gastrointestinal tract in rats. REGULATORY PEPTIDES 2003; 115:91-9. [PMID: 12972324 DOI: 10.1016/s0167-0115(03)00145-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Peptides of the trefoil factor family (TFF1, TFF2 and TFF3) are co-secreted with mucus in most organ systems and are believed to interact with mucins to produce high-viscosity, stable gel complexes. We have previously demonstrated that cells in the GI tract possess binding sites to TFF2 and that injected TFF2 ends up in the mucus layer. In the present study, tissue binding and metabolism of parenterally administered human TFF1 and TFF3 in rats were described and compared to the immunohistochemical localization of the TFF peptides. 125I-TFF1 monomer and 125I-TFF3 mono- and dimer were given intravenously to female Wistar rats. The tissue distribution was assessed by gamma counting of organ samples and by autoradiography of histological sections. The degradation of 125I-TFF3 was studied by means of trichloracetic acid (TCA) precipitation and the saturability of the binding by administration of excess unlabelled peptide. The TFF peptides were localized in histologic sections from the GI tract by immunohistochemistry. Injected TFF3 dimer (12%) was taken up by the GI tract. At autoradiography, grains were localized to the same cells that were immunoreactive to TFF2. The binding could be displaced by excess TFF3. Similar binding was observed for the TFF1 and TFF3 monomers apart from binding in the stomach, where the uptake was only 15% in comparison to the dimer. There was no specific binding outside the GI tract and no binding to TFF1 or TFF3 immunoreactive cells. In conclusion, the TFF2-binding cells in the gastrointestinal tract seem to have basolateral, receptor-like activity to all three TFF peptides. The mucous neck cells of the stomach predominantly take up TFFs with two trefoil domains, indicating a different receptor-like activity in the stomach compared to the rest of the GI tract.
Collapse
Affiliation(s)
- S S Poulsen
- Anatomy Department B, The Panum Institute, University of Copenhagen, 3 Blegdamsvej, 2200 N, Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
28
|
Chen YH, Lu Y, De Plaen IG, Wang LY, Tan XD. Transcription factor NF-kappaB signals antianoikic function of trefoil factor 3 on intestinal epithelial cells. Biochem Biophys Res Commun 2000; 274:576-82. [PMID: 10924320 DOI: 10.1006/bbrc.2000.3176] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Transcription factor NF-kappaB has both pro-apoptotic and anti-apoptotic properties depending on the cell type. Its role in the intestinal epithelial cell has not been well elucidated. Trefoil factor 3 (TFF3) is an anti-apoptotic peptide secreted by intestinal goblet cells. Here we show that TFF3 activated NF-kappaB p50/p65 heterodimer within 1 h in IEC-18 cells (a nontransformed rat intestinal epithelial cell line). Moreover, we found that TFF3-treated IEC-18 cells are resistant to anoikis, an anchorage-related apoptosis in epithelium. In addition, the stable expression of a mutant form of the endogenous NF-kappaB inhibitor (IkappaBalpha(mut)) in IEC-18 cells results in a significant attenuation of anti-anoikic effect of TFF3. Taken together, these data indicate that (1) TFF3 is an endogenous gastrointestinal peptide with anti-anoikic property; (2) TFF3 activates NF-kappaB in enterocytes; and (3) TFF3-induced resistance to anoikis in intestinal epithelial cells is mediated by a distinct signaling cascade linked to NF-kappaB. Furthermore, our study implicates NF-kappaB as an important regulator in survival pathway of intestinal epithelial cells.
Collapse
Affiliation(s)
- Y H Chen
- Department of Pathology, Northwestern University, Chicago, Illinois, 60614, USA
| | | | | | | | | |
Collapse
|
29
|
Kinoshita K, Taupin DR, Itoh H, Podolsky DK. Distinct pathways of cell migration and antiapoptotic response to epithelial injury: structure-function analysis of human intestinal trefoil factor. Mol Cell Biol 2000; 20:4680-90. [PMID: 10848594 PMCID: PMC85884 DOI: 10.1128/mcb.20.13.4680-4690.2000] [Citation(s) in RCA: 147] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The trefoil peptide intestinal trefoil factor (ITF) plays a critical role in the protection of colonic mucosa and is essential to restitution after epithelial damage. These functional properties are accomplished through coordinated promotion of cell migration and inhibition of apoptosis. ITF contains a unique three-looped trefoil motif formed by intrachain disulfide bonds among six conserved cysteine residues, which is thought to contribute to its marked protease resistance. ITF also has a seventh cysteine residue, which permits homodimer formation. A series of cysteine-to-serine substitutions and a C-terminally truncated ITF were made by PCR site-directed mutagenesis. Any alteration of the trefoil motif or truncation resulted in loss of protease resistance. However, neither an intact trefoil domain nor dimerization was required to promote cell migration. This pro-restitution activity correlated with the ability of the ITF mutants to activate mitogen-activated protein (MAP) kinase independent of phosphorylation of the epidermal growth factor (EGF) receptor. In contrast, only intact ITF retained both phosphatidylinositol 3-kinase and the EGF receptor-dependent antiapoptotic effect in HCT116 and IEC-6 cells. The inability to block apoptosis correlated with a loss of trefoil peptide-induced transactivation of the EGF receptor or Akt kinase in HT-29 cells. In addition to defining structural requirements for the functional properties of ITF, these findings demonstrate that distinct intracellular signaling pathways mediate the effects of ITF on cell migration and apoptosis.
Collapse
Affiliation(s)
- K Kinoshita
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | |
Collapse
|
30
|
Thim L, Mørtz E. Isolation and characterization of putative trefoil peptide receptors. REGULATORY PEPTIDES 2000; 90:61-8. [PMID: 10828494 DOI: 10.1016/s0167-0115(00)00110-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Mammalian trefoil factors (TFFs) constitute a group of three peptides (TFF1, TFF2 and TFF3) widely distributed in the gastrointestinal tract. Although a mucosal protection/healing effect of these peptides is well documented the mechanism of action is still unknown. A mucosal membrane extract was prepared from porcine stomach scrapings and incubated with a gel containing immobilized porcine TFF2. The affinity gel material was specifically eluted with a neutral buffer containing a high concentration of the ligand (porcine TFF2). A subsequent SDS-gel electrophoresis showed one protein with a MW of approximately 220 kDa and three proteins with MW around 140 kDa. The proteins were analyzed by trypsin digestion followed by mass spectrometric sequencing of tryptic fragments. In this way a 140-kDa beta subunit of fibronectin receptor and a 224-kDa CRP-Ductin gene product were identified. The CRP-Ductin gene product (also named MUCLIN), which is expressed in the intestinal crypts, is characterized by being a membrane protein with a short cytoplasmic region, a transmembrane domain and a large extracellular region. This protein thus fulfils some of the criteria for being a TFF receptor or a TFF binding protein.
Collapse
Affiliation(s)
- L Thim
- Department of Protein Chemistry, Health Care Discovery, Novo Nordisk A/S, DK-2880, Bagsvaerd, Denmark.
| | | |
Collapse
|
31
|
Tan XD, Chen YH, Liu QP, Gonzalez-Crussi F, Liu XL. Prostanoids mediate the protective effect of trefoil factor 3 in oxidant-induced intestinal epithelial cell injury: role of cyclooxygenase-2. J Cell Sci 2000; 113 ( Pt 12):2149-55. [PMID: 10825288 DOI: 10.1242/jcs.113.12.2149] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Trefoil factors are small peptides found in several mammalian tissues including gut, respiratory tract and brain. Their physiological function is not well understood. Among them, trefoil factor 3 (intestinal trefoil factor) is known to be cytoprotective in the gut. However, the molecular mechanism and secondary mediators of trefoil factor 3 action are not known. In the present study, we examined whether the cyclooxygenase pathway is involved in trefoil factor 3 action. We showed that trefoil factor 3 significantly induces the production of prostaglandin E(2) and prostaglandin I(2) in IEC-18 cells (an intestinal epithelial cell line) in a dose dependent manner. Western blot and immunohistochemistry revealed that trefoil factor 3 (2.5 microM) up-regulates the expression of cyclooxygenase-2 but not cyclooxygenase-1 in IEC-18 cells. Treating cells with trefoil factor 3 (10 microM) significantly attenuated reactive oxygen species-induced IEC-18 cell injury. This effect is blocked by NS-398 (10 microM), a selective cyclooxygenase-2 inhibitor. Moreover, we demonstrated that exogenously administered carbacyclin (1 microM, a stable analogue of prostaglandin I(2)) and/or prostaglandin E(2) (1 microM) caused a significant reduction of reactive oxygen species-induced cell injury, mimicking the effect of trefoil factor 3. In summary, our results indicate that trefoil factor 3 activates cyclooxygenase-2 in intestinal epithelium to produce prostaglandin I(2) and prostaglandin E(2), which function as survival factors and mediate the cytoprotective action of trefoil factor 3 against oxidant injury.
Collapse
Affiliation(s)
- X D Tan
- Department of Pathology, Children's Memorial Hospital, Northwestern University, Chicago, IL 60614, USA.
| | | | | | | | | |
Collapse
|
32
|
Jagla W, Wiede A, Dietzmann K, Rutkowski K, Hoffmann W. Co-localization of TFF3 peptide and oxytocin in the human hypothalamus. FASEB J 2000; 14:1126-31. [PMID: 10834934 DOI: 10.1096/fasebj.14.9.1126] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
TFF-peptides (formerly P domain peptides, trefoil factors) are typical secretory products of many mucous epithelial cells. TFF3 is also synthesized in the hypothalamus and has anxiolytic or anxiogenic activities when injected into the rat amygdala. Here we show by immunohistochemistry that TFF3 is localized to a distinct population of neurons of the human hypothalamic paraventricular and supraoptic nuclei. Generally, TFF3-positive cells are co-localized in oxytocin-producing cells and not in vasopressin-producing cells. Relatively large amounts of TFF3-but not TFF1 and TFF2-are present in the posterior lobe of the human pituitary, where it is probably released into the bloodstream. Furthermore, TFF3 was also detectable in human postmortem cerebrospinal fluid.
Collapse
Affiliation(s)
- W Jagla
- Institut für Molekularbiologie und Medizinische Chemie, Otto-von-Guericke-Universität, D-39120 Magdeburg, Germany
| | | | | | | | | |
Collapse
|
33
|
Tomasetto C, Masson R, Linares JL, Wendling C, Lefebvre O, Chenard MP, Rio MC. pS2/TFF1 interacts directly with the VWFC cysteine-rich domains of mucins. Gastroenterology 2000; 118:70-80. [PMID: 10611155 DOI: 10.1016/s0016-5085(00)70415-x] [Citation(s) in RCA: 133] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Trefoil factors (TFFs) are secreted gastrointestinal proteins that have been shown to protect and promote healing of the gastrointestinal tract. Moreover, pS2/TFF1 is essential for normal differentiation of the gastric mucosa because deficient mice develop antropyloric adenomas. To date, it is unclear how TFFs mediate their functions. METHODS Using the yeast 2-hybrid system, we attempted to identify murine TFF1 interacting proteins by screening a stomach and duodenum complementary DNA (cDNA) expression library. RESULTS Four positive clones were isolated. Sequence and expression studies showed that they corresponded to the murine counterpart of human cDNA sequences encoding carboxy-terminal fragments of mMuc2 (489 residues) and mMuc5AC (427, 430, and 894 residues) mucin proteins. Mutagenesis experiments showed that TFF1 interacts with the 2 mucins through binding with their VWFC1 and VWFC2 (von Willebrand factor C) cysteine-rich domains. CONCLUSIONS These results show that the gastrointestinal protective effect of TFF1, and presumably of the other TFFs, is caused at least partially by their participation, via mucin binding, in the correct organization of the mucous layer that protects the apical side of the mucosa from deleterious luminal agents.
Collapse
Affiliation(s)
- C Tomasetto
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM Unité 184/ULP, Illkirch, France
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
BACKGROUND Trefoil peptides are a family of small proteins that are expressed in a site-specific fashion by certain epithelial tissues. These peptides appear to be important in mucosal healing processes and in neoplastic disease. METHODS This manuscript reviews the relevant literature obtained by an extensive text word search of the Medline database and a manual search of references from the articles identified. RESULTS AND CONCLUSION Trefoil peptides are aberrantly expressed by a wide range of human carcinomas and gastrointestinal inflammatory conditions. They impart protection from injury to the gastrointestinal mucosa by possible interaction with mucin glycoproteins. Trefoil peptides influence epithelial cell migration and mucosal restitution following injury. In the future, serum levels of trefoil peptides might be used as markers for both neoplastic and inflammatory diseases. In addition, novel therapies based on such peptides might be used for gastrointestinal inflammatory conditions and to accelerate repair of the gastrointestinal mucosa after surgery.
Collapse
Affiliation(s)
- R J Longman
- University Department of Surgery, Bristol Royal Infirmary, UK
| | | | | |
Collapse
|
35
|
Intestinal trefoil factor binds to intestinal epithelial cells and induces nitric oxide production: priming and enhancing effects of mucin. Biochem J 1999. [PMID: 10051448 DOI: 10.1042/0264-6021:3380745] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Intestinal trefoil factor (ITF or TFF3), NO and epithelium-associated mucin have important roles in sustaining mucosal integrity in the gastrointestinal tract. In the present study we examined ITF-binding molecules on IEC-18 cells (an intestinal epithelial cell line) with the use of flow cytometry and localized these molecules on the cell surface by confocal microscopy. Furthermore, we studied the interaction of mucin and ITF and their co-operative effect on NO production by the epithelium. Stimulation of cells with mucin (5 mg/ml) for 90 min resulted in a 5-fold increase in ITF binding. Treatment of IEC-18 cells with actinomycin D or cycloheximide attenuated mucin-enhanced ITF binding. Ligand blot analysis confirmed the induction of ITF-binding protein in IEC-18 cells by mucin. These results indicate that transcriptional and translational mechanisms are involved in the effect of mucin. Treatment with ITF overnight resulted in a low level of nitrite production by the cells, a 5-fold increase over control, in a concentration-dependent manner. ITF-induced NO production was attenuated by 1400W, a selective type II nitric oxide synthase (NOS2) inhibitor. By immunoblotting we found that NOS2 was up-regulated by ITF treatment. Priming IEC-18 cells with mucin for 90 min enhanced the effect of ITF on NO production, suggesting that the up-regulation of ITF-binding molecules by mucin might be physiologically relevant. Taken together, these observations indicate (1) that ITF-binding molecules that are up-regulated by mucin exist on the intestinal epithelial surface, and (2) that ITF modulates epithelial NO production via the NOS2 pathway, which is enhanced by mucin.
Collapse
|
36
|
Intestinal trefoil factor binds to intestinal epithelial cells and induces nitric oxide production: priming and enhancing effects of mucin. Biochem J 1999. [PMID: 10051448 DOI: 10.1042/bj3380745] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Intestinal trefoil factor (ITF or TFF3), NO and epithelium-associated mucin have important roles in sustaining mucosal integrity in the gastrointestinal tract. In the present study we examined ITF-binding molecules on IEC-18 cells (an intestinal epithelial cell line) with the use of flow cytometry and localized these molecules on the cell surface by confocal microscopy. Furthermore, we studied the interaction of mucin and ITF and their co-operative effect on NO production by the epithelium. Stimulation of cells with mucin (5 mg/ml) for 90 min resulted in a 5-fold increase in ITF binding. Treatment of IEC-18 cells with actinomycin D or cycloheximide attenuated mucin-enhanced ITF binding. Ligand blot analysis confirmed the induction of ITF-binding protein in IEC-18 cells by mucin. These results indicate that transcriptional and translational mechanisms are involved in the effect of mucin. Treatment with ITF overnight resulted in a low level of nitrite production by the cells, a 5-fold increase over control, in a concentration-dependent manner. ITF-induced NO production was attenuated by 1400W, a selective type II nitric oxide synthase (NOS2) inhibitor. By immunoblotting we found that NOS2 was up-regulated by ITF treatment. Priming IEC-18 cells with mucin for 90 min enhanced the effect of ITF on NO production, suggesting that the up-regulation of ITF-binding molecules by mucin might be physiologically relevant. Taken together, these observations indicate (1) that ITF-binding molecules that are up-regulated by mucin exist on the intestinal epithelial surface, and (2) that ITF modulates epithelial NO production via the NOS2 pathway, which is enhanced by mucin.
Collapse
|
37
|
Abstract
The pathogenesis of both ulcerative colitis and Crohn's disease is unknown but these forms of inflammatory bowel disease (IBD) may be associated with an inability of the intestinal mucosa to protect itself from luminal challenges and/or inappropriate repair following intestinal injury. Numerous cell populations regulate these broad processes through the expression of a complex array of peptides and other agents. Growth factors can be distinguished by their actions regulating cell proliferation. These factors also mediate processes such as extracellular matrix formation, cell migration and differentiation, immune regulation, and tissue remodeling. Several families of growth factors may play an important role in IBD including: epidermal growth factor family (EGF) [transforming growth factor alpha (TGF alpha), EGF itself, and others], the transforming growth factor beta (TGF beta) super family, insulin-like growth factors (IGF), fibroblast growth factors (FGF), hepatocyte growth factor (HGF), trefoil factors, platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF) and others. Collectively these families may determine susceptibility of IBD mucosa to injury and facilitate tissue repair.
Collapse
Affiliation(s)
- P L Beck
- Department of Medicine, Massachusetts General Hospital 02114, USA
| | | |
Collapse
|
38
|
Abstract
The pathogenesis of both ulcerative colitis and Crohn's disease is unknown but these forms of inflammatory bowel disease (IBD) may be associated with an inability of the intestinal mucosa to protect itself from luminal challenges and/or inappropriate repair following intestinal injury. Numerous cell populations regulate these broad processes through the expression of a complex array of peptides and other agents. Growth factors can be distinguished by their actions regulating cell proliferation. These factors also mediate processes such as extracellular matrix formation, cell migration and differentiation, immune regulation, and tissue remodeling. Several families of growth factors may play an important role in IBD including: epidermal growth factor family (EGF) [transforming growth factor alpha (TGF alpha), EGF itself, and others], the transforming growth factor beta (TGF beta) super family, insulin-like growth factors (IGF), fibroblast growth factors (FGF), hepatocyte growth factor (HGF), trefoil factors, platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF) and others. Collectively these families may determine susceptibility of IBD mucosa to injury and facilitate tissue repair.
Collapse
Affiliation(s)
- P L Beck
- Department of Medicine, Massachusetts General Hospital 02114, USA
| | | |
Collapse
|
39
|
Schwarzberg H, Kalbacher H, Hoffmann W. Differential behavioral effects of TFF peptides: injections of synthetic TFF3 into the rat amygdala. Pharmacol Biochem Behav 1999; 62:173-8. [PMID: 9972861 DOI: 10.1016/s0091-3057(98)00137-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
TFF peptides (formerly named P-domain peptides or trefoil factors) are also released from the brain as well as being secreted typically by mucin producing cells. The amygdala, besides the hypothalamus, represents a defined neuronal locality of TFF3 synthesis. In a passive avoidance test synthetic TFF3/monomer or 0.9% sodium chloride (control) was injected bilaterally into the basolateral nucleus of the amygdala of male rats either immediately (consolidation test) or 23 h after a footshock (retrieval test). Application of a low TFF3 dose (2 x 6 pg) decreased avoidance latency in a time dependent manner. In contrast, a high dose (2 x 60 pg) increased avoidance latency. Maximal effects of TFF3 were observed about 24 h after the injection. This bidirectional effect was also observed using the elevated plus-maze test. The locomotor activity on the open arms was significantly increased 24 h after a low dose injection of TFF3 into the amygdala. In contrast, a high-dose injection significantly decreased the activity on the open arms. The results of both tests can be explained by an anxiolytic effect at a low dose and an anxiogenic effect at a high dose of synthetic TFF3/monomer.
Collapse
Affiliation(s)
- H Schwarzberg
- Institut für Neurophysiologie, Otto-von-Guericke-Universität, Magdeburg, Germany
| | | | | |
Collapse
|
40
|
Efstathiou JA, Pignatelli M. Modulation of epithelial cell adhesion in gastrointestinal homeostasis. THE AMERICAN JOURNAL OF PATHOLOGY 1998; 153:341-7. [PMID: 9708793 PMCID: PMC1852965 DOI: 10.1016/s0002-9440(10)65576-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/02/1998] [Indexed: 01/09/2023]
Affiliation(s)
- J A Efstathiou
- Cancer and Immunogenetics Laboratory, Imperial Cancer Research Fund, Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | | |
Collapse
|
41
|
Efstathiou JA, Noda M, Rowan A, Dixon C, Chinery R, Jawhari A, Hattori T, Wright NA, Bodmer WF, Pignatelli M. Intestinal trefoil factor controls the expression of the adenomatous polyposis coli-catenin and the E-cadherin-catenin complexes in human colon carcinoma cells. Proc Natl Acad Sci U S A 1998; 95:3122-7. [PMID: 9501226 PMCID: PMC19705 DOI: 10.1073/pnas.95.6.3122] [Citation(s) in RCA: 122] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/1997] [Indexed: 02/06/2023] Open
Abstract
Intestinal trefoil factor 3 (TFF3) is a member of the trefoil family of peptides, small molecules constitutively expressed in epithelial tissues, including the gastrointestinal tract. TFF3 has been shown to promote migration of intestinal epithelial cells in vitro and to enhance mucosal healing and epithelial restitution in vivo. In this study, we evaluated the effect of recombinant TFF3 (rTFF3) stimulation on the expression and cellular localization of the epithelial (E)-cadherin-catenin complex, a prime mediator of Ca2+ dependent cell-cell adhesion, and the adenomatous polyposis coli (APC)-catenin complex in HT29, HCT116, and SW480 colorectal carcinoma cell lines. Stimulation by rTFF3 (10(-9) M and 10(-8) M) for 20-24 hr led to cell detachment and to a reduction in intercellular adhesion in HT29 and HCT116 cells. In both cell lines, E-cadherin expression was down-regulated. The expression of APC, alpha-catenin and beta-catenin also was decreased in HT29 cells, with a translocation of APC into the nucleus. No change in either cell adhesion or in the expression of E-cadherin, the catenins, and APC was detected in SW480 cells. In addition, TFF3 induced DNA fragmentation and morphological changes characteristic of apoptosis in HT29. Tyrphostin, a competitive inhibitor of protein tyrosine kinases, inhibited the effects of TFF3. Our results indicate that by perturbing the complexes between E-cadherin, beta-catenin, and associated proteins, TFF3 may modulate epithelial cell adhesion, migration, and survival.
Collapse
Affiliation(s)
- J A Efstathiou
- Division of Investigative Science, Imperial College of Science, Technology and Medicine, Hammersmith Campus, Du Cane Road, London W12 ONN, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|