1
|
Yang X, Ye T, Rong L, Peng H, Tong J, Xiao X, Wan X, Guo J. GATA4 Forms a Positive Feedback Loop with CDX2 to Transactivate MUC2 in Bile Acids-Induced Gastric Intestinal Metaplasia. Gut Liver 2024; 18:414-425. [PMID: 36860162 PMCID: PMC11096910 DOI: 10.5009/gnl220394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 03/03/2023] Open
Abstract
Background/Aims Gastric intestinal metaplasia (GIM), a common precancerous lesion of gastric cancer, can be caused by bile acid reflux. GATA binding protein 4 (GATA4) is an intestinal transcription factor involved in the progression of gastric cancer. However, the expression and regulation of GATA4 in GIM has not been clarified. Methods The expression of GATA4 in bile acid-induced cell models and human specimens was examined. The transcriptional regulation of GATA4 was investigated by chromatin immunoprecipitation and luciferase reporter gene analysis. An animal model of duodenogastric reflux was used to confirm the regulation of GATA4 and its target genes by bile acids. Results GATA4 expression was elevated in bile acid-induced GIM and human specimens. GATA4 bound to the promoter of mucin 2 (MUC2) and stimulate its transcription. GATA4 and MUC2 expression was positively correlated in GIM tissues. Nuclear transcription factor-κB activation was required for the upregulation of GATA4 and MUC2 in bile acid-induced GIM cell models. GATA4 and caudal-related homeobox 2 (CDX2) reciprocally transactivated each other to drive the transcription of MUC2. In chenodeoxycholic acid-treated mice, MUC2, CDX2, GATA4, p50, and p65 expression levels were increased in the gastric mucosa. Conclusions GATA4 is upregulated and can form a positive feedback loop with CDX2 to transactivate MUC2 in GIM. NF-κB signaling is involved in the upregulation of GATA4 by chenodeoxycholic acid.
Collapse
Affiliation(s)
- Xiaofang Yang
- Department of Gastroenterology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
| | - Ting Ye
- Department of Gastroenterology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
| | - Li Rong
- Department of Gastroenterology, Bishan Hospital of Chongqing, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Peng
- Department of Gastroenterology, Bishan Hospital of Chongqing, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Tong
- Department of Gastroenterology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
| | - Xiao Xiao
- Department of Gastroenterology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
| | - Xiaoqiang Wan
- Department of Gastroenterology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
| | - Jinjun Guo
- Department of Gastroenterology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University School of Medicine, Chongqing, China
- Department of Gastroenterology, Bishan Hospital of Chongqing, Bishan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Akinsuyi OS, Xhumari J, Ojeda A, Roesch LFW. Gut permeability among Astronauts during Space missions. LIFE SCIENCES IN SPACE RESEARCH 2024; 41:171-180. [PMID: 38670644 DOI: 10.1016/j.lssr.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/02/2024] [Accepted: 03/13/2024] [Indexed: 04/28/2024]
Abstract
The space environment poses substantial challenges to human physiology, including potential disruptions in gastrointestinal health. Gut permeability has only recently become widely acknowledged for its potential to cause adverse effects on a systemic level, rendering it a critical factor to investigate in the context of spaceflight. Here, we propose that astronauts experience the onset of leaky gut during space missions supported by transcriptomic and metagenomic analysis of human and murine samples. A genetic map contributing to intestinal permeability was constructed from a systematic review of current literature. This was referenced against our re-analysis of three independent transcriptomic datasets which revealed significant changes in gene expression patterns associated with the gut barrier. Specifically, in astronauts during flight, we observed a substantial reduction in the expression genes that are crucial for intestinal barrier function, goblet cell development, gut microbiota modulation, and immune responses. Among rodent spaceflight studies, differential expression of cytokines, chemokines, and genes which regulate mucin production and post-translational modifications suggest a similar dysfunction of intestinal permeability. Metagenomic analysis of feces from two murine studies revealed a notable reduction probiotic, short chain fatty acid-producing bacteria and an increase in the Gram-negative pathogens, including Citrobacter rodentium, Enterobacter cloacea, Klebsiella aerogenes, and Proteus hauseri which promote LPS circulation, a recipe for barrier disruption and systemic inflammatory activation. These findings emphasize the critical need to understand the underlying mechanisms and develop interventions to maintain gastrointestinal health in space.
Collapse
Affiliation(s)
- Oluwamayowa S Akinsuyi
- Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville 32611, FL, USA
| | - Jessica Xhumari
- Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville 32611, FL, USA
| | - Amanda Ojeda
- Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville 32611, FL, USA
| | - Luiz F W Roesch
- Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville 32611, FL, USA.
| |
Collapse
|
3
|
Kim HW, Lee SY, Hur SJ, Kil DY, Kim JH. Effects of functional nutrients on chicken intestinal epithelial cells induced with oxidative stress. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2023; 65:1040-1052. [PMID: 37969347 PMCID: PMC10640939 DOI: 10.5187/jast.2023.e22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 11/17/2023]
Abstract
The objective of this study was to investigate the protective effects of functional nutrients including various functional amino acids, vitamins, and minerals on chicken intestinal epithelial cells (cIECs) treated with oxidative stress. The cIECs were isolated from specific pathogen free eggs. Cells were exposed to 0 mM supplement (control), 20 mM threonine (Thr), 0.4 mM tryptophan (Trp), 1 mM glycine (Gly), 10 μM vitamin C (VC), 40 μM vitamin E (VE), 5 μM vitamin A (VA), 34 μM chromium (Cr), 0.42 μM selenium (Se), and 50 μM zinc (Zn) for 24 h with 6 replicates for each treatment. After 24 h, cells were further incubated with fresh culture medium (positive control, PC) or 1 mM H2O2 with different supplements (negative control, NC and each treatment). Oxidative stress was measured by cell proliferation, whereas tight junction barrier function was analyzed by fluorescein isothiocyanate (FITC)-dextran permeability and transepithelial electrical resistance (TEER). Results indicated that cell viability and TEER values were less (p < 0.05) in NC treatments with oxidative stress than in PC treatments. In addition, FITC-dextran values were greater (p < 0.05) in NC treatments with oxidative stress than in PC treatments. The supplementations of Thr, Trp, Gly, VC, and VE in cells treated with H2O2 showed greater (p < 0.05) cell viability than the supplementation of VA, Cr, Se, and Zn. The supplementations of Trp, Gly, VC, and Se in cells treated with H2O2 showed the least (p < 0.05) cellular permeability. In addition, the supplementation of Thr, VE, VA, Cr, and Zn in cells treated with H2O2 decreased (p < 0.05) cellular permeability. At 48 h, the supplementations of Thr, Trp, and Gly in cells treated with H2O2 showed the greatest (p < 0.05) TEER values among all treatments, and the supplementations of VC and VE in cells treated with H2O2 showed greater (p < 0.05) TEER values than the supplementations of VA, Cr, Se, and Zn in cells treated with H2O2. In conclusion, Thr, Trp, Gly, and VC supplements were effective in improving cell viability and intestinal barrier function of cIECs exposed to oxidative stress.
Collapse
Affiliation(s)
- Hyun Woo Kim
- Department of Animal Science and
Technology, Chung-Ang University, Anseong 17546, Korea
| | - Seung Yun Lee
- Department of Animal Science (BK21 Four),
Institute of Agriculture Life Science, Gyeongsang National
University, Jinju 52725, Korea
| | - Sun Jin Hur
- Department of Animal Science and
Technology, Chung-Ang University, Anseong 17546, Korea
| | - Dong Yong Kil
- Department of Animal Science and
Technology, Chung-Ang University, Anseong 17546, Korea
| | - Jong Hyuk Kim
- Department of Animal Science, Chungbuk
National University, Cheongju 28644, Korea
| |
Collapse
|
4
|
Nascimento CM, Casaro MC, Perez ER, Ribeiro WR, Mayer MPA, Ishikawa KH, Lino-dos-Santos-Franco A, Pereira JNB, Ferreira CM. Experimental allergic airway inflammation impacts gut homeostasis in mice. Heliyon 2023; 9:e16429. [PMID: 37484240 PMCID: PMC10360590 DOI: 10.1016/j.heliyon.2023.e16429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/30/2023] [Accepted: 05/16/2023] [Indexed: 07/25/2023] Open
Abstract
Background /Aims: Epidemiological data show that there is an important relationship between respiratory and intestinal diseases. To improve our understanding on the interconnectedness between the lung and intestinal mucosa and the overlap between respiratory and intestinal diseases, our aim was to investigate the influence of ovalbumin (OVA)-induced allergic airway inflammation on gut homeostasis. Methods A/J mice were sensitized and challenged with OVA. The animals were euthanized 24 h after the last challenge, lung inflammation was determined by evaluating cells in Bronchoalveolar lavage fluid, serum anti-OVA IgG titers and colon morphology, inflammation and integrity of the intestinal mucosa were investigated. IL-4 and IL-13 levels and myeloperoxidase activity were determined in the colon samples. The expression of genes involved in inflammation and mucin production at the gut mucosa was also evaluated. Results OVA challenge resulted not only in lung inflammation but also in macroscopic alterations in the gut such as colon shortening, increased myeloperoxidase activity and loss of integrity in the colonic mucosal. Neutral mucin intensity was lower in the OVA group, which was followed by down-regulation of transcription of ATOH1 and up-regulation of TJP1 and MUC2. In addition, the OVA group had higher levels of IL-13 and IL-4 in the colon. Ova-specific IgG1 and OVA-specific IgG2a titers were higher in the serum of the OVA group than in controls. Conclusions Our data using the OVA experimental model suggested that challenges in the respiratory system may result not only in allergic airway inflammation but also in the loss of gut homeostasis.
Collapse
Affiliation(s)
- Carolina Martins Nascimento
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, Universidade Federal de São Paulo, Diadema, SP, Brazil
| | - Mateus Campos Casaro
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, Universidade Federal de São Paulo, Diadema, SP, Brazil
| | - Evelyn Roxana Perez
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, Universidade Federal de São Paulo, Diadema, SP, Brazil
| | - Willian Rodrigues Ribeiro
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, Universidade Federal de São Paulo, Diadema, SP, Brazil
| | - Marcia Pinto Alves Mayer
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Karin Hitomi Ishikawa
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | | | - Caroline Marcantonio Ferreira
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, Universidade Federal de São Paulo, Diadema, SP, Brazil
| |
Collapse
|
5
|
Yang S, Yu M. Role of Goblet Cells in Intestinal Barrier and Mucosal Immunity. J Inflamm Res 2021; 14:3171-3183. [PMID: 34285541 PMCID: PMC8286120 DOI: 10.2147/jir.s318327] [Citation(s) in RCA: 165] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/29/2021] [Indexed: 12/17/2022] Open
Abstract
Goblet cells and the mucus they secrete serve as an important barrier, preventing pathogens from invading the mucosa to cause intestinal inflammation. The perspective regarding goblet cells and mucus has changed, with current evidence suggesting that they are not passive but play a positive role in maintaining intestinal tract immunity and mucosal homeostasis. Goblet cells could obtain luminal antigens, presenting them to the underlying antigen-presenting cells (APCs) that induces adaptive immune responses. Various immunomodulatory factors can promote the differentiation and maturation of goblet cells, and the secretion of mucin. The abnormal proliferation and differentiation of goblet cells, as well as the deficiency synthesis and secretion of mucins, result in intestinal mucosal barrier dysfunction. This review provides an extensive outline of the signaling pathways that regulate goblet cell proliferation and differentiation and control mucins synthesis and secretion to elucidate how altering these pathways affects goblet functionality. Furthermore, the interaction between mucins and goblet cells in intestinal mucosal immunology is described. Therefore, the contribution of goblet cells and mucus in promoting gut defense and homeostasis is illustrated, while clarifying the regulatory mechanisms involved may allow the development of new therapeutic strategies for intestinal disorders.
Collapse
Affiliation(s)
- Songwei Yang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, 400030, People's Republic of China
| | - Min Yu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| |
Collapse
|
6
|
Characterization of the regulatory 5'-flanking region of bovine mucin 2 (MUC2) gene. Mol Cell Biochem 2021; 476:2847-2856. [PMID: 33730299 DOI: 10.1007/s11010-021-04133-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 03/06/2021] [Indexed: 10/21/2022]
Abstract
Throughout the intestinal epithelium surface there is an intricate polymer network composed by gel-forming mucins, which plays a protective role due to the formation of a physical, chemical and immunological barrier between the organism and the environment. Mucin 2 (MUC2) is the main mucin in the small and large intestine, and it is expressed specifically in the gastrointestinal tract (GIT), which makes its promoter region an important candidate for expression of heterologous genes of biotechnological interest in the GIT of bovine and other ruminants. In order to characterize the bovine MUC2 promoter we designed primers to amplify and isolate a candidate region for this promoter. The amplified sequence was confirmed by sequencing and cloned into a plasmid vector containing the luciferase (LUC) reporter gene. The regulatory sites of the MUC2 promoter already described in the literature were used to find the putative regulatory sites in the bovine MUC2 promoter region. With these data, some deletions were performed in order to find the promoter sequence with greatest expression capacity and specificity. The constructions were tested by transient transfection assays in LoVo cells (human colorectal adenocarcinoma) and bovine fibroblasts. The quantification of the relative expression of the promoter was measured using dual-luciferase assays. Real-time PCR was performed to analyze the expression of endogenous MUC2. The results presented herein prove that the isolated sequence corresponds to the promoter of bovine MUC2 gene, since it was able to induce expression of a reporter gene in an in vitro cell culture experimental platform.
Collapse
|
7
|
Nanoencapsulation (in vitro and in vivo) as an efficient technology to boost the potential of garlic essential oil as alternatives for antibiotics in broiler nutrition. Animal 2020; 15:100022. [PMID: 33573947 DOI: 10.1016/j.animal.2020.100022] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 11/23/2022] Open
Abstract
The addition of essential oil (EO) as chitosan encapsulated can increase the efficiency of these oils in broiler feeding. Therefore, the objective of the current research was to explore the antibacterial and antioxidant potential of garlic essential oil (GEO) (free vs. nanoencapsulated) and their effects on performance, gene expression of mucin2, microbial, and morphology of intestine in broilers. A total of 900 1-day-old male broilers (Ross 308) were assigned to six dietary treatments (0, 100, and 200 mg/kg free GEO and 0 (contain of chitosan), 100 and 200 mg/kg nanoencapsulated GEO) with a 2 × 3 factorial arrangement based on completely randomized design. Garlic essential oil encapsulation with chitosan significantly enhanced antibacterial and antioxidant parameters. At 100 mg/kg nanoencapsulated GEO had significant (P < 0.01) advantages in improving BW gain (BWG) (22-42 and 0-42) and feed conversion ratio (FCR) (0-42). Maximum feed intake (FI) was also associated with the control group (P < 0.05). Broilers fed on 100 mg/kg of nanoencapsulated GEO showed higher villi length and width relative to other treatments and villi length to crypt depth ratio as well (P < 0.01). The nanoencapsulation process of GEO (P < 0.01) affected the Lactobacilli population in the digesta of ileo-caecum and mucin2 gene expression. In broiler chickens, the tested EO, especially nanoencapsulated type, enhanced more evaluated parameters. Because of its ideal properties, nanoencasulating with chitosan may also be an effective and inexpensive way to protect bioactive compounds and improve GEO effects in broiler chickens.
Collapse
|
8
|
Pothuraju R, Krishn SR, Gautam SK, Pai P, Ganguly K, Chaudhary S, Rachagani S, Kaur S, Batra SK. Mechanistic and Functional Shades of Mucins and Associated Glycans in Colon Cancer. Cancers (Basel) 2020; 12:E649. [PMID: 32168759 PMCID: PMC7139953 DOI: 10.3390/cancers12030649] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 02/08/2023] Open
Abstract
Mucus serves as the chief protective barrier against pathogenic and mechanical insults in respiratory, gastrointestinal, and urogenital tracts. Altered mucin expression, the major component of mucus, in conjunction with differential glycosylation has been strongly associated with both benign and malignant pathologies of colon. Mucins and their associated glycans arbitrate their impact sterically as well as mechanically by altering molecular and microbial spectrum during pathogenesis. Mucin expression in normal and pathological conditions is regulated by nonspecific (dietary factors and gut microbiota) and specific (epigenetic and transcriptional) modulators. Further, recent studies highlight the impact of altering mucin glycome (cancer-associated carbohydrate antigens including Tn, Sialyl-Tn, Sialyl-Lew A, and Sialyl-Lewis X) on host immunomodulation, antitumor immunity, as well as gut microbiota. In light of emerging literature, the present review article digs into the impact of structural organization and of expressional and glycosylation alteration of mucin family members on benign and malignant pathologies of colorectal cancer.
Collapse
Affiliation(s)
- Ramesh Pothuraju
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.P.); (S.R.K.); (S.K.G.); (P.P.); (K.G.); (S.C.); (S.R.); (S.K.)
| | - Shiv Ram Krishn
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.P.); (S.R.K.); (S.K.G.); (P.P.); (K.G.); (S.C.); (S.R.); (S.K.)
| | - Shailendra K. Gautam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.P.); (S.R.K.); (S.K.G.); (P.P.); (K.G.); (S.C.); (S.R.); (S.K.)
| | - Priya Pai
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.P.); (S.R.K.); (S.K.G.); (P.P.); (K.G.); (S.C.); (S.R.); (S.K.)
| | - Koelina Ganguly
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.P.); (S.R.K.); (S.K.G.); (P.P.); (K.G.); (S.C.); (S.R.); (S.K.)
| | - Sanjib Chaudhary
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.P.); (S.R.K.); (S.K.G.); (P.P.); (K.G.); (S.C.); (S.R.); (S.K.)
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.P.); (S.R.K.); (S.K.G.); (P.P.); (K.G.); (S.C.); (S.R.); (S.K.)
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.P.); (S.R.K.); (S.K.G.); (P.P.); (K.G.); (S.C.); (S.R.); (S.K.)
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.P.); (S.R.K.); (S.K.G.); (P.P.); (K.G.); (S.C.); (S.R.); (S.K.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68105, USA
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
9
|
Reynolds IS, Fichtner M, McNamara DA, Kay EW, Prehn JHM, Burke JP. Mucin glycoproteins block apoptosis; promote invasion, proliferation, and migration; and cause chemoresistance through diverse pathways in epithelial cancers. Cancer Metastasis Rev 2020; 38:237-257. [PMID: 30680581 DOI: 10.1007/s10555-019-09781-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Overexpression of mucin glycoproteins has been demonstrated in many epithelial-derived cancers. The significance of this overexpression remains uncertain. The aim of this paper was to define the association of mucin glycoproteins with apoptosis, cell growth, invasion, migration, adhesion, and clonogenicity in vitro as well as tumor growth, tumorigenicity, and metastasis in vivo in epithelial-derived cancers by performing a systematic review of all published data. A systematic review of PubMed, Embase, and the Cochrane Central Register of Controlled Trials was performed to identify all papers that evaluated the association between mucin glycoproteins with apoptosis, cell growth, invasion, migration, adhesion, and clonogenicity in vitro as well as tumor growth, tumorigenicity, and metastasis in vivo in epithelial-derived cancers. PRISMA guidelines were adhered to. Results of individual studies were extracted and pooled together based on the organ in which the cancer was derived from. The initial search revealed 2031 papers, of which 90 were deemed eligible for inclusion in the study. The studies included details on MUC1, MUC2, MUC4, MUC5AC, MUC5B, MUC13, and MUC16. The majority of studies evaluated MUC1. MUC1 overexpression was consistently associated with resistance to apoptosis and resistance to chemotherapy. There was also evidence that overexpression of MUC2, MUC4, MUC5AC, MUC5B, MUC13, and MUC16 conferred resistance to apoptosis in epithelial-derived cancers. The overexpression of mucin glycoproteins is associated with resistance to apoptosis in numerous epithelial cancers. They cause resistance through diverse signaling pathways. Targeting the expression of mucin glycoproteins represents a potential therapeutic target in the treatment of epithelial-derived cancers.
Collapse
Affiliation(s)
- Ian S Reynolds
- Department of Colorectal Surgery, Beaumont Hospital, Dublin 9, Ireland
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - Michael Fichtner
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - Deborah A McNamara
- Department of Colorectal Surgery, Beaumont Hospital, Dublin 9, Ireland
- Department of Surgery, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - Elaine W Kay
- Department of Pathology, Beaumont Hospital, Dublin 9, Ireland
- Department of Pathology, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - Jochen H M Prehn
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - John P Burke
- Department of Colorectal Surgery, Beaumont Hospital, Dublin 9, Ireland.
| |
Collapse
|
10
|
Straub D, Oude Elferink RPJ, Jansen PLM, Bergman JJGHM, Parikh K, Krishnadath KK. Glyco-conjugated bile acids drive the initial metaplastic gland formation from multi-layered glands through crypt-fission in a murine model. PLoS One 2019; 14:e0220050. [PMID: 31348796 PMCID: PMC6660124 DOI: 10.1371/journal.pone.0220050] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 07/08/2019] [Indexed: 12/22/2022] Open
Abstract
Bile acid reflux is known to be associated with the development of Barrett’s esophagus and esophageal adenocarcinoma (EAC), yet the role of specific bile acids and the mechanism behind the metaplastic changes is unclear. Here, we demonstrate that multi-layered glandular structures at the squamo-columnar junction in mice contain multiple cell lineages, which resemble the human esophageal submucosal gland ducts. Exposing mice to patient’s refluxates induced expansion of multi-layered glandular structures and development of columnar metaplasia at the squamo-columnar junction. The glycine conjugated bile acids induced an intestinal type of metaplasia more typical for Barrett’s esophagus. Through lineage tracing, we excluded the involvement of K5+, DCLK1+, and LGR5+ progenitor cells as the primary source in the development of the glandular metaplastic epithelium. We show that the mechanism behind development of metaplasia involves crypt fission and may be independent of stem cell proliferation. Our findings support the hypothesis that in humans, BE arises from non-squamous cells residing in submucosal gland ducts and that induction of intestinal type of metaplasia is most effectively induced by glycine-conjugated bile acids. These novel insights may lead to more effective strategies to prevent development of Barrett’s esophagus and esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Danielle Straub
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, Amsterdam, The Netherlands
| | | | - Peter L. M. Jansen
- Department of Gastrointestinal and Liver Disease, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Kaushal Parikh
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Kausilia K. Krishnadath
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, Amsterdam, The Netherlands
- Department of Gastroenterology, Academic Medical Center, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
11
|
Podany A, Rauchut J, Wu T, Kawasawa YI, Wright J, Lamendella R, Soybel DI, Kelleher SL. Excess Dietary Zinc Intake in Neonatal Mice Causes Oxidative Stress and Alters Intestinal Host-Microbe Interactions. Mol Nutr Food Res 2018; 63:e1800947. [PMID: 30513548 DOI: 10.1002/mnfr.201800947] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 10/23/2018] [Indexed: 12/11/2022]
Abstract
SCOPE Greater than 68% of young infants are exposed to dietary zinc (Zn) levels that are higher than the Tolerable Upper Intake Limit. However, the consequences of excess dietary Zn during early life on intestinal function and host-microbe interactions are unknown. METHODS AND RESULTS Neonatal mice are gavaged with 100 Zn µg d-1 from postnatal day (PN) 2 through PN10 and indices of intestinal function and host-microbe interactions are compared to unsupplemented mice. Excess dietary Zn causes oxidative stress, increases goblet cell number and mucus production, and are associated with increased intestinal permeability and systemic inflammation. Over 900 genes are differentially expressed; 413 genes display a fold-change >1.60. The Gene Ontology Biological processes most significantly affected include biological adhesion, the immune system, metabolic processes, and response to stimulus. Key genes most highly and significantly upregulated include ALDH2, MT1, TMEM6, CDK20, and COX62b, while CALU, ST3GAL4, CRTC2, SLC28A2, and COMMA1 are downregulated. These changes are associated with a microbiome enriched in pathogenic taxa including Pseudomonadales and Campylobacter, and greater expression of bacterial stress response genes. CONCLUSION Excess dietary Zn may have unforeseen influences on epithelial signaling pathways, barrier function, and luminal ecology in the intestine that may have long-term consequences on intestinal health.
Collapse
Affiliation(s)
- Abigail Podany
- Department of Surgery, Penn State Hershey College of Medicine, Hershey, PA
| | - Jessica Rauchut
- Department of Surgery, Penn State Hershey College of Medicine, Hershey, PA
| | - Tong Wu
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA
| | - Yuka Imamura Kawasawa
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA.,Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, Penn State Hershey College of Medicine, Hershey, PA
| | - Justin Wright
- Department of Biology, Juniata College, Huntingdon, PA
| | | | - David I Soybel
- Department of Surgery, Penn State Hershey College of Medicine, Hershey, PA
| | - Shannon L Kelleher
- Department of Surgery, Penn State Hershey College of Medicine, Hershey, PA.,Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA.,Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, PA
| |
Collapse
|
12
|
Yamashita MSDA, Melo EO. Mucin 2 (MUC2) promoter characterization: an overview. Cell Tissue Res 2018; 374:455-463. [PMID: 30218241 DOI: 10.1007/s00441-018-2916-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 08/13/2018] [Indexed: 12/24/2022]
Abstract
Transgenic livestock have been studied with a well-known interest in improving quantitative and qualitative traits. In order to direct heterologous gene expression, it is indispensable to identify and characterize a promoter suitable for directing the expression of the gene of interest (GOI) in a tissue-specific way. The gastrointestinal tract is a desirable target for gene expression in several mammalian models. Throughout the surface of the intestinal epithelium, there is an intricate polymer network, formed by gel-forming mucins (especially MUC2 and MUC5AC, of which MUC2 is the major one), which plays a protective role due to the formation of a physical, chemical and immunological barrier between the organism and the environment. The characterization of the gel-forming mucins is difficult because of their large size and repetitive DNA sequences and domains. The main mucin in the small and large intestine, mucin 2 (MUC2), is expressed specifically in goblet cells. MUC2 plays an important role in intestinal homeostasis and its disruption is associated with several diseases and carcinomas. This mucin is also an important marker for elucidating mechanisms that regulate differentiation of the secretory cell lineage. This review presents the state of the art of MUC2 promoter structure and functional characterization.
Collapse
Affiliation(s)
| | - Eduardo O Melo
- EMBRAPA Genetic Resources and Biotechnology, PqEB Av W5 Norte, Brasilia, DF, 70770-917, Brazil
| |
Collapse
|
13
|
TGF-βRII Knock-down in Pancreatic Cancer Cells Promotes Tumor Growth and Gemcitabine Resistance. Importance of STAT3 Phosphorylation on S727. Cancers (Basel) 2018; 10:cancers10080254. [PMID: 30065235 PMCID: PMC6116183 DOI: 10.3390/cancers10080254] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 01/05/2023] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is one of the most deadly cancers in the Western world because of a lack of early diagnostic markers and efficient therapeutics. At the time of diagnosis, more than 80% of patients have metastasis or locally advanced cancer and are therefore not eligible for surgical resection. Pancreatic cancer cells also harbour a high resistance to chemotherapeutic drugs such as gemcitabine that is one of the main palliative treatments for PDAC. Proteins involved in TGF-β signaling pathway (SMAD4 or TGF-βRII) are frequently mutated in PDAC (50–80%). TGF-β signalling pathway plays antagonistic roles during carcinogenesis by initially inhibiting epithelial growth and later promoting the progression of advanced tumors and thus emerged as both tumor suppressor and oncogenic pathways. In order to decipher the role of TGF-β in pancreatic carcinogenesis and chemoresistance, we generated CAPAN-1 and CAPAN-2 cell lines knocked down for TGF-βRII (first actor of TGF-β signaling). The impact on biological properties of these TGF-βRII-KD cells was studied both in vitro and in vivo. We show that TGF-βRII silencing alters tumor growth and migration as well as resistance to gemcitabine. TGF-βRII silencing also leads to S727 STAT3 and S63 c-Jun phosphorylation, decrease of MRP3 and increase of MRP4 ABC transporter expression and induction of a partial EMT phenotype. These markers associated with TGF-β signaling pathways may thus appear as potent therapeutic tools to better treat/manage pancreatic cancer.
Collapse
|
14
|
Lee S, Kelleher SL. Molecular regulation of lactation: The complex and requisite roles for zinc. Arch Biochem Biophys 2016; 611:86-92. [DOI: 10.1016/j.abb.2016.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/10/2016] [Accepted: 04/04/2016] [Indexed: 12/22/2022]
|
15
|
The oncogenic receptor ErbB2 modulates gemcitabine and irinotecan/SN-38 chemoresistance of human pancreatic cancer cells via hCNT1 transporter and multidrug-resistance associated protein MRP-2. Oncotarget 2016; 6:10853-67. [PMID: 25890497 PMCID: PMC4484424 DOI: 10.18632/oncotarget.3414] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/19/2015] [Indexed: 02/06/2023] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is one of the most deadly cancers because of a lack of early diagnotic markers and efficient therapeutics. The fluorinated analog of deoxycytidine, gemcitabine and emerging FOLFIRINOX protocol (5-fluorouracil (5-FU), irinotecan/SN-38, oxaliplatin and leucovorin) are the main chemotherapies to treat PDAC. The ErbB2/HER2 oncogenic receptor is commonly overexpressed in PDAC. In this context, we aimed to decipher the ErbB2-mediated mechanisms of chemoresistance to the two main chemotherapy protocols used to treat PDAC. ErbB2 knocking down (KD) in CAPAN-1 and CAPAN-2 cells led to an increased sensitivity to gemcitabine and an increased resistance to irinotecan/SN-38 both in vitro and in vivo (subcuteanous xenografts) This was correlated to an increase of hCNT1 and hCNT3 transporters and ABCG2, MRP1 and MRP2 ATP-binding cassette transporters expression and resistance to cell death. We also show that MRP2 is repressed following activation of JNK, Erk1/2 and NF-κB pathways by ErbB2. Finally, in datasets of human PDAC samples, ErbB2 and MRP2 expression was conversely correlated. Altogether, we propose that ErbB2 mediates several intracellular mechanisms linked to PDAC cell chemoresistance that may represent potential targets in order to ameliorate chemotherapy response and allow stratification of patients eligible for either gemcitabine or FOLFIRINOX treatment.
Collapse
|
16
|
Ferretti VA, Segal-Eiras A, Barbeito CG, Croce MV. Temporal and spatial expression of Muc2 and Muc5ac mucins during rat respiratory and digestive tracts development. Res Vet Sci 2016; 104:136-45. [PMID: 26850552 DOI: 10.1016/j.rvsc.2015.12.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/15/2015] [Accepted: 12/22/2015] [Indexed: 10/22/2022]
Abstract
Secreted mucins constitute a crucial part of the gel that protects respiratory and digestive epithelia, being MUC2/Muc2 the predominant gel-forming mucin of the intestine while MUC5AC/Muc5ac is one of the gel-forming mucins most expressed at the airways. In this study, we have analyzed Muc2 and Muc5ac during rat development by using immunohistochemistry, Western blotting and RT-PCR. We demonstrated that rat Muc2 was expressed in fetal intestinal goblet cells of surface epithelium of villi and developing Lieberkühn crypts. In neonates and adults, Muc2 was expressed at luminal goblet cells of small and large intestine and at gastric mucous and glandular cells. Muc5ac protein was observed in embryonic gastric and lung samples; expression increased during development and postnatal and adult life. After birth, a low reaction was detected at the tracheal surface epithelium and glands, which increased in adults.
Collapse
Affiliation(s)
- V A Ferretti
- Centre of Basic and Applied Immunological Research, Faculty of Medical Sciences, National University of La Plata, La Plata, Argentina
| | - A Segal-Eiras
- Centre of Basic and Applied Immunological Research, Faculty of Medical Sciences, National University of La Plata, La Plata, Argentina
| | - C G Barbeito
- Laboratory of Histology and Embryology, Faculty of Veterinary Sciences, National University of La Plata, Argentina
| | - M V Croce
- Centre of Basic and Applied Immunological Research, Faculty of Medical Sciences, National University of La Plata, La Plata, Argentina.
| |
Collapse
|
17
|
Tréhoux S, Lahdaoui F, Delpu Y, Renaud F, Leteurtre E, Torrisani J, Jonckheere N, Van Seuningen I. Micro-RNAs miR-29a and miR-330-5p function as tumor suppressors by targeting the MUC1 mucin in pancreatic cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2392-403. [PMID: 26036346 DOI: 10.1016/j.bbamcr.2015.05.033] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 05/11/2015] [Accepted: 05/28/2015] [Indexed: 12/14/2022]
Abstract
MUC1 is an oncogenic mucin overexpressed in several epithelial cancers, including pancreatic ductal adenocarcinoma, and is considered as a potent target for cancer therapy. To control cancer progression, miRNAs became very recently, major targets and tools to inhibit oncogene expression. Inhibiting MUC1 using miRNAs appears thus as an attractive strategy to reduce cancer progression. However, potent miRNAs and associated mechanisms regulating MUC1 expression remain to be identified. To this aim, we undertook to study MUC1 regulation by miRNAs in pancreatic cancer cells and identify those with tumor suppressive activity. MiRNAs potentially targeting the 3'-UTR, the coding region, or the 5'-UTR of MUC1 were selected using an in silico approach. Our in vitro and in vivo experiments indicate that miR-29a and miR-330-5p are strong inhibitors of MUC1 expression in pancreatic cancer cells through direct binding to MUC1 3'-UTR. MUC1 regulation by the other selected miRNAs (miR-183, miR-200a, miR-876-3p and miR-939) was found to be indirect. MiR-29a and miR-330-5p are also deregulated in human pancreatic cancer cell lines and tissues and in pancreatic tissues of Kras(G12D) mice. In vitro, miR-29a and miR-330-5p inhibit cell proliferation, cell migration, cell invasion and sensitize pancreatic cancer cells to gemcitabine. In vivo intra-tumoral injection of these two miRNAs in xenografted pancreatic tumors led to reduced tumor growth. Altogether, we have identified miR-29a and miR-330-5p as two new tumor suppressive miRNAs that inhibit the expression of MUC1 oncogenic mucin in pancreatic cancer cells.
Collapse
Affiliation(s)
- Solange Tréhoux
- Inserm, UMR-S1172, Jean-Pierre Aubert Research Center, Team "Mucins, Epithelial Differentiation and Carcinogenesis", Rue Polonovski, 59045 Lille cedex, France; Université de Lille 2, 42 rue Paul Duez, 59000 Lille, France; Centre Hospitalier Régional et Universitaire de Lille, 59037 Lille cedex, France
| | - Fatima Lahdaoui
- Inserm, UMR-S1172, Jean-Pierre Aubert Research Center, Team "Mucins, Epithelial Differentiation and Carcinogenesis", Rue Polonovski, 59045 Lille cedex, France; Université de Lille 2, 42 rue Paul Duez, 59000 Lille, France; Centre Hospitalier Régional et Universitaire de Lille, 59037 Lille cedex, France
| | - Yannick Delpu
- Inserm, UMR1037, Cancer Research Center of Toulouse, 1 avenue Jean Poulhes, 31432 Toulouse cedex 4, France
| | - Florence Renaud
- Inserm, UMR-S1172, Jean-Pierre Aubert Research Center, Team "Mucins, Epithelial Differentiation and Carcinogenesis", Rue Polonovski, 59045 Lille cedex, France; Université de Lille 2, 42 rue Paul Duez, 59000 Lille, France; Centre Hospitalier Régional et Universitaire de Lille, 59037 Lille cedex, France; Institut de Pathologie, Centre de Biologie Pathologie, Boulevard du Professeur Jules Leclercq, 59037 Lille, France
| | - Emmanuelle Leteurtre
- Inserm, UMR-S1172, Jean-Pierre Aubert Research Center, Team "Mucins, Epithelial Differentiation and Carcinogenesis", Rue Polonovski, 59045 Lille cedex, France; Université de Lille 2, 42 rue Paul Duez, 59000 Lille, France; Centre Hospitalier Régional et Universitaire de Lille, 59037 Lille cedex, France; Institut de Pathologie, Centre de Biologie Pathologie, Boulevard du Professeur Jules Leclercq, 59037 Lille, France
| | - Jérôme Torrisani
- Inserm, UMR1037, Cancer Research Center of Toulouse, 1 avenue Jean Poulhes, 31432 Toulouse cedex 4, France
| | - Nicolas Jonckheere
- Inserm, UMR-S1172, Jean-Pierre Aubert Research Center, Team "Mucins, Epithelial Differentiation and Carcinogenesis", Rue Polonovski, 59045 Lille cedex, France; Université de Lille 2, 42 rue Paul Duez, 59000 Lille, France; Centre Hospitalier Régional et Universitaire de Lille, 59037 Lille cedex, France
| | - Isabelle Van Seuningen
- Inserm, UMR-S1172, Jean-Pierre Aubert Research Center, Team "Mucins, Epithelial Differentiation and Carcinogenesis", Rue Polonovski, 59045 Lille cedex, France; Université de Lille 2, 42 rue Paul Duez, 59000 Lille, France; Centre Hospitalier Régional et Universitaire de Lille, 59037 Lille cedex, France.
| |
Collapse
|
18
|
|
19
|
Gronnier C, Bruyère E, Lahdaoui F, Jonckheere N, Perrais M, Leteurtre E, Piessen G, Mariette C, Van Seuningen I. The MUC1 mucin regulates the tumorigenic properties of human esophageal adenocarcinomatous cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2432-7. [PMID: 25003315 DOI: 10.1016/j.bbamcr.2014.06.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/19/2014] [Accepted: 06/27/2014] [Indexed: 02/07/2023]
Abstract
MUC1 is a membrane-bound mucin known to participate in tumor proliferation. It has been shown that MUC1 pattern of expression is modified during esophageal carcinogenesis, with a progressive increase from metaplasia to adenocarcinoma. The principal cause of development of esophageal adenocarcinoma is gastro-esophageal reflux and MUC1 was previously shown to be up-regulated by several bile acids present in reflux. In this report, our aim was thus to determine whether MUC1 plays a role in biological properties of human esophageal cancer cells. For that, a stable MUC1-deficient esophageal cancer cell line was established using a shRNA approach. In vitro (proliferation, migration and invasion) and in vivo (tumor growth following subcutaneous xenografts in SCID mice) biological properties of MUC1-deficient cells were analyzed. Our results show that esophageal cancer cells lacking MUC1 were less proliferative and had decreased migration and invasion properties. These alterations were accompanied by a decreased activity of NFKB p65, Akt and MAPK (p44/42, JNK and p38) pathways. MCM6 and TSG101 tumor-associated markers were also decreased. Subcutaneous xenografts showed a significant decrease in tumor size when cells did not express MUC1. Altogether, the data indicate that MUC1 plays a key role in proliferative, migrating and invasive properties of esophageal cancer cells as well as in tumor growth promotion. MUC1 mucin appears thus as a good therapeutic target to slow down esophageal tumor progression.
Collapse
Affiliation(s)
- Caroline Gronnier
- Inserm, UMR837, Jean-Pierre Aubert Research Center, Team 5 "Mucins, Epithelial Differentiation and Carcinogenesis", Lille, France; Université Lille-Nord de France, Lille, France; Department of Digestive and Oncological Surgery, University Hospital Claude Huriez, Lille, France
| | - Emilie Bruyère
- Inserm, UMR837, Jean-Pierre Aubert Research Center, Team 5 "Mucins, Epithelial Differentiation and Carcinogenesis", Lille, France; Université Lille-Nord de France, Lille, France; Department of Digestive and Oncological Surgery, University Hospital Claude Huriez, Lille, France
| | - Fatima Lahdaoui
- Inserm, UMR837, Jean-Pierre Aubert Research Center, Team 5 "Mucins, Epithelial Differentiation and Carcinogenesis", Lille, France; Université Lille-Nord de France, Lille, France; Department of Digestive and Oncological Surgery, University Hospital Claude Huriez, Lille, France
| | - Nicolas Jonckheere
- Inserm, UMR837, Jean-Pierre Aubert Research Center, Team 5 "Mucins, Epithelial Differentiation and Carcinogenesis", Lille, France; Université Lille-Nord de France, Lille, France
| | - Michaël Perrais
- Inserm, UMR837, Jean-Pierre Aubert Research Center, Team 5 "Mucins, Epithelial Differentiation and Carcinogenesis", Lille, France; Université Lille-Nord de France, Lille, France
| | - Emmanuelle Leteurtre
- Inserm, UMR837, Jean-Pierre Aubert Research Center, Team 5 "Mucins, Epithelial Differentiation and Carcinogenesis", Lille, France; Université Lille-Nord de France, Lille, France; Centre de Biologie-Pathologie, Department of Pathology, Centre Hospitalier Régional et Universitaire, Lille, France
| | - Guillaume Piessen
- Inserm, UMR837, Jean-Pierre Aubert Research Center, Team 5 "Mucins, Epithelial Differentiation and Carcinogenesis", Lille, France; Université Lille-Nord de France, Lille, France; Department of Digestive and Oncological Surgery, University Hospital Claude Huriez, Lille, France
| | - Christophe Mariette
- Inserm, UMR837, Jean-Pierre Aubert Research Center, Team 5 "Mucins, Epithelial Differentiation and Carcinogenesis", Lille, France; Université Lille-Nord de France, Lille, France; Department of Digestive and Oncological Surgery, University Hospital Claude Huriez, Lille, France
| | - Isabelle Van Seuningen
- Inserm, UMR837, Jean-Pierre Aubert Research Center, Team 5 "Mucins, Epithelial Differentiation and Carcinogenesis", Lille, France; Université Lille-Nord de France, Lille, France.
| |
Collapse
|
20
|
Lahdaoui F, Delpu Y, Vincent A, Renaud F, Messager M, Duchêne B, Leteurtre E, Mariette C, Torrisani J, Jonckheere N, Van Seuningen I. miR-219-1-3p is a negative regulator of the mucin MUC4 expression and is a tumor suppressor in pancreatic cancer. Oncogene 2014; 34:780-8. [PMID: 24608432 DOI: 10.1038/onc.2014.11] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 11/29/2013] [Accepted: 01/01/2014] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the most lethal cancers in the world with one of the worst outcome. The oncogenic mucin MUC4 has been identified as an actor of pancreatic carcinogenesis as it is involved in many processes regulating pancreatic cancer cell biology. MUC4 is not expressed in healthy pancreas whereas it is expressed very early in pancreatic carcinogenesis. Targeting MUC4 in these early steps may thus appear as a promising strategy to slow-down pancreatic tumorigenesis. miRNA negative regulation of MUC4 could be one mechanism to efficiently downregulate MUC4 gene expression in early pancreatic neoplastic lesions. Using in silico studies, we found two putative binding sites for miR-219-1-3p in the 3'-UTR of MUC4 and showed that miR-219-1-3p expression is downregulated both in PDAC-derived cell lines and human PDAC tissues compared with their normal counterparts. We then showed that miR-219-1-3p negatively regulates MUC4 mucin expression via its direct binding to MUC4 3'-UTR. MiR-219-1-3p overexpression (transient and stable) in pancreatic cancer cell lines induced a decrease of cell proliferation associated with a decrease of cyclin D1 and a decrease of Akt and Erk pathway activation. MiR-219-1-3p overexpression also decreased cell migration. Furthermore, miR-219-1-3p expression was found to be conversely correlated with Muc4 expression in early pancreatic intraepithelial neoplasia lesions of Pdx1-Cre;LSL-Kras(G12D) mice. Most interestingly, in vivo studies showed that miR-219-1-3p injection in xenografted pancreatic tumors in mice decreased both tumor growth and MUC4 mucin expression. Altogether, these results identify miR-219-1-3p as a new negative regulator of MUC4 oncomucin that possesses tumor-suppressor activity in PDAC.
Collapse
Affiliation(s)
- F Lahdaoui
- 1] Inserm, UMR837, Jean Pierre Aubert Research Center (JPARC), Team 5 'Mucins, epithelial differentiation and carcinogenesis', rue Polonovski, Lille Cedex, France [2] Université Lille Nord de France, Lille, France [3] Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - Y Delpu
- 1] Inserm, UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France [2] Université Paul Sabatier, Toulouse, France
| | - A Vincent
- 1] Inserm, UMR837, Jean Pierre Aubert Research Center (JPARC), Team 5 'Mucins, epithelial differentiation and carcinogenesis', rue Polonovski, Lille Cedex, France [2] Université Lille Nord de France, Lille, France [3] Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - F Renaud
- 1] Inserm, UMR837, Jean Pierre Aubert Research Center (JPARC), Team 5 'Mucins, epithelial differentiation and carcinogenesis', rue Polonovski, Lille Cedex, France [2] Université Lille Nord de France, Lille, France [3] Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - M Messager
- 1] Inserm, UMR837, Jean Pierre Aubert Research Center (JPARC), Team 5 'Mucins, epithelial differentiation and carcinogenesis', rue Polonovski, Lille Cedex, France [2] Université Lille Nord de France, Lille, France [3] Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - B Duchêne
- 1] Inserm, UMR837, Jean Pierre Aubert Research Center (JPARC), Team 5 'Mucins, epithelial differentiation and carcinogenesis', rue Polonovski, Lille Cedex, France [2] Université Lille Nord de France, Lille, France [3] Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - E Leteurtre
- 1] Inserm, UMR837, Jean Pierre Aubert Research Center (JPARC), Team 5 'Mucins, epithelial differentiation and carcinogenesis', rue Polonovski, Lille Cedex, France [2] Université Lille Nord de France, Lille, France [3] Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - C Mariette
- 1] Inserm, UMR837, Jean Pierre Aubert Research Center (JPARC), Team 5 'Mucins, epithelial differentiation and carcinogenesis', rue Polonovski, Lille Cedex, France [2] Université Lille Nord de France, Lille, France [3] Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - J Torrisani
- 1] Inserm, UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France [2] Université Paul Sabatier, Toulouse, France
| | - N Jonckheere
- 1] Inserm, UMR837, Jean Pierre Aubert Research Center (JPARC), Team 5 'Mucins, epithelial differentiation and carcinogenesis', rue Polonovski, Lille Cedex, France [2] Université Lille Nord de France, Lille, France [3] Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - I Van Seuningen
- 1] Inserm, UMR837, Jean Pierre Aubert Research Center (JPARC), Team 5 'Mucins, epithelial differentiation and carcinogenesis', rue Polonovski, Lille Cedex, France [2] Université Lille Nord de France, Lille, France [3] Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| |
Collapse
|
21
|
Effect of dietary zinc oxide on morphological characteristics, mucin composition and gene expression in the colon of weaned piglets. PLoS One 2014; 9:e91091. [PMID: 24609095 PMCID: PMC3946750 DOI: 10.1371/journal.pone.0091091] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 02/07/2014] [Indexed: 12/17/2022] Open
Abstract
The trace element zinc is often used in the diet of weaned piglets, as high doses have resulted in positive effects on intestinal health. However, the majority of previous studies evaluated zinc supplementations for a short period only and focused on the small intestine. The hypothesis of the present study was that low, medium and high levels of dietary zinc (57, 164 and 2,425 mg Zn/kg from zinc oxide) would affect colonic morphology and innate host defense mechanisms across 4 weeks post-weaning. Histological examinations were conducted regarding the colonic morphology and neutral, acidic, sialylated and sulphated mucins. The mRNA expression levels of mucin (MUC) 1, 2, 13, 20, toll-like receptor (TLR) 2, 4, interleukin (IL)-1β, 8, 10, interferon-γ (IFN-γ) and transforming growth factor-β (TGF-β) were also measured. The colonic crypt area increased in an age-depending manner, and the greatest area was found with medium concentration of dietary zinc. With the high concentration of dietary zinc, the number of goblet cells containing mixed neutral-acidic mucins and total mucins increased. Sialomucin containing goblet cells increased age-dependently. The expression of MUC2 increased with age and reached the highest level at 47 days of age. The expression levels of TLR2 and 4 decreased with age. The mRNA expression of TLR4 and the pro-inflammatory cytokine IL-8 were down-regulated with high dietary zinc treatment, while piglets fed with medium dietary zinc had the highest expression. It is concluded that dietary zinc level had a clear impact on colonic morphology, mucin profiles and immunological traits in piglets after weaning. Those changes might support local defense mechanisms and affect colonic physiology and contribute to the reported reduction of post-weaning diarrhea.
Collapse
|
22
|
Gronnier C, Bruyère E, Piessen G, Briez N, Bot J, Buob D, Leteurtre E, Van Seuningen I, Mariette C. Operatively induced chronic reflux in rats: a suitable model for studying esophageal carcinogenesis? Surgery 2013; 154:955-67. [PMID: 24084597 DOI: 10.1016/j.surg.2013.05.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 05/16/2013] [Indexed: 12/15/2022]
Abstract
BACKGROUND The mechanisms of esophageal reflux leading to esophageal adenocarcinoma (EA) remain poorly understood. This study appraises critically an operatively induced chronic reflux rat model. METHODS We randomized 108 Sprague-Dawley rats into 2 experimental groups; one was performing esophagoduodenal (ED) anastomosis with or without gastrectomy to induce duodeno-esophageal reflux (DER group; n = 63), and the other involved duodeno-gastro-esophageal reflux (DGER group; n = 45). Control groups included (i) Roux-en-Y esophagojejunal anastomosis, (ii) laparotomy alone, (iii) subtotal gastrectomy to induce duodenogastric reflux (DGR group), and (iv) the same procedure as in the DGER group plus proton pump inhibition (PPI group). The esophagus underwent histologic and molecular analyses. RESULTS The prevalence of Barrett's esophagus (BE), dysplasia, and EA in the experimental groups was 41%, 7%, and 11%, respectively. Histologic and molecular analyses in groups DER, DGER, and DGR suggested that BE occurred through de novo intestinal metaplasia and proximal migration of duodenal cells. No distant metastases were identified. The molecular characteristics of both BE and EA were similar to humans. BE was more common, and dysplasia and EA less frequent in the DER group when compared with the DGER group (44% vs 24% [P = .038] and 7% vs 25% [P = .012], respectively). Compared with the DGER group, carcinogenic sequence occurred less frequently in the PPI-treated group (P = .019). CONCLUSION Despite pathophysiologic differences with humans, the rat model of esophagoduodenostomy reproduces accurately histologic and molecular lesions in the carcinogenetic sequence of BE and allowed us to identify novel, tumor-associated proteins that may be potential biomarkers and new therapeutic targets in EA.
Collapse
Affiliation(s)
- Caroline Gronnier
- Inserm, UMR837, Jean-Pierre Aubert Research Center, Team 5 "Mucins, epithelial differentiation and carcinogenesis", Lille, France; Université Lille Nord de France, Lille, France; Department of Digestive and Oncological Surgery, University Hospital Claude Huriez, Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Castro IC, Breiling A, Luetkenhaus K, Ceteci F, Hausmann S, Kress S, Lyko F, Rudel T, Rapp UR. MYC-induced epigenetic activation of GATA4 in lung adenocarcinoma. Mol Cancer Res 2012; 11:161-72. [PMID: 23239811 DOI: 10.1158/1541-7786.mcr-12-0414-t] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Human lung cancer is a disease with high incidence and accounts for most cancer-related deaths in both men and women. Metastasis is a common event in non-small cell lung carcinoma (NSCLC), diminishing the survival chance of the patients with this type of tumor. It has been shown that MYC is involved in the development of metastasis from NSCLC, but the mechanism underlying this switch remained to be identified. Here, we focus on GATA4 as a MYC target in the development of metastasis with origin in lung adenocarcinoma, the most common type of NSCLC. Epigenetic alterations at the GATA4 promoter level were observed after MYC expression in lung adenocarcinoma in vivo and in vitro. Such alterations include site-specific demethylation that accompanies the displacement of the MYC-associated zinc finger protein (MAZ) from the GATA4 promoter, which leads to GATA4 expression. Histone modification analysis of the GATA4 promoter revealed a switch from repressive histone marks to active histone marks after MYC binding, which corresponds to active GATA4 expression. Our results thus identify a novel epigenetic mechanism by which MYC activates GATA4 leading to metastasis in lung adenocarcinoma, suggesting novel potential targets for the development of antimetastatic therapy.
Collapse
Affiliation(s)
- Inês C Castro
- Biocenter, Department of Microbiology, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ya-Feng Z, Gang S, Xiao-Tong Z, Zhi-Qi Z, Xia-Jing L, Song-Bo W, Li-Na W, Yong-Liang Z, Qing-Yan J. Identification of an intestine-specific promoter and inducible expression of bacterial α-galactosidase in mammalian cells by a lac operon system. J Anim Sci Biotechnol 2012; 3:32. [PMID: 23111091 PMCID: PMC3527164 DOI: 10.1186/2049-1891-3-32] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Accepted: 10/15/2012] [Indexed: 12/04/2022] Open
Abstract
Background α-galactosidase has been widely used in animal husbandry to reduce anti-nutritional factors (such as α-galactoside) in feed. Intestine-specific and substrate inducible expression of α-galactosidase would be highly beneficial for transgenic animal production. Methods To achieve the intestine-specific and substrate inducible expression of α-galactosidase, we first identified intestine-specific promoters by comparing the transcriptional activity and tissue specificity of four intestine-specific promoters from human intestinal fatty acid binding protein, rat intestinal fatty acid binding protein, human mucin-2 and human lysozyme. We made two chimeric constructs combining the promoter and enhancer of human mucin-2, rat intestinal trefoil factor and human sucrase-isomaltase. Then a modified lac operon system was constructed to investigate the induction of α-galactosidase expression and enzyme activity by isopropyl β-D-1-thiogalactopyranoside (IPTG) and an α-galactosidase substrate, α-lactose. We declared that the research carried out on human (Zhai Yafeng) was in compliance with the Helsinki Declaration, and experimental research on animals also followed internationally recognized guidelines. Results The activity of the human mucin-2 promoter was about 2 to 3 times higher than that of other intestine-specific promoters. In the lac operon system, the repressor significantly decreased (P < 0.05) luciferase activity by approximately 6.5-fold and reduced the percentage of cells expressing green fluorescent protein (GFP) by approximately 2-fold. In addition, the expression level of α-galactosidase mRNA was decreased by 6-fold and α-galactosidase activity was reduced by 8-fold. In line with our expectations, IPTG and α-lactose supplementation reversed (P < 0.05) the inhibition and produced a 5-fold increase of luciferase activity, an 11-fold enhancement in the percentage of cells with GFP expression and an increase in α-galactosidase mRNA abundance (by about 5-fold) and α-galactosidase activity (by about 7-fold). Conclusions We have successfully constructed a high specificity inducible lac operon system in an intestine-derived cell line, which could be of great value for gene therapy applications and transgenic animal production.
Collapse
Affiliation(s)
- Zhai Ya-Feng
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Shigeshiro M, Tanabe S, Suzuki T. Repeated exposure to water immersion stress reduces the Muc2 gene level in the rat colon via two distinct mechanisms. Brain Behav Immun 2012; 26:1061-5. [PMID: 22683765 DOI: 10.1016/j.bbi.2012.05.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 05/24/2012] [Accepted: 05/25/2012] [Indexed: 12/13/2022] Open
Abstract
The mucin family plays a number of important roles in intestinal homeostasis. Among its members, the gel-forming Muc2, produced in goblet cells, is a major component of mucus and contributes to intestinal barrier integrity. Whereas psychological stress is known to impair intestinal barrier, the effects of chronic or repeated stress on mucin expression and goblet cell differentiation have not been well documented. The present study first examined the effects of different levels of exposure (3 days, 1 and 2 weeks) to water immersion stress on intestinal mucin gene expression in rats, and then explored the mechanisms underlying the stress-induced decrease in the colonic Muc2 level. Repeated water immersion stress for 1 and 2 weeks decreased colonic Muc2 gene levels to 40% of that of non-stressed animals, while exposure to stress for 3 days induced only a 25% decrease. The goblet cell numbers counts in the colons of the 1- and 2-week stress groups, but not the 3-day stress group, were decreased to 85% of that in non-stressed animals. Cdx2 expression, a transcriptional factor related to Muc2 synthesis in the goblet cells, was decreased in all stress groups, whereas Rath1 and Klf4 expressions, transcriptional factors related to goblet cell differentiation in Notch signaling were decreased in the 1-week stress group. Collectively, the repeated exposure to water immersion stress decreases Muc2 synthesis in the goblet cells via decreased Cdx2 expression and subsequently reduces the goblet cell number via Notch signaling suppression.
Collapse
Affiliation(s)
- Mizuki Shigeshiro
- Department of Biofunctional Science and Technology, Graduate School of Biosphere Science, Hiroshima University, Japan
| | | | | |
Collapse
|
26
|
GATA-4/-6 and HNF-1/-4 families of transcription factors control the transcriptional regulation of the murine Muc5ac mucin during stomach development and in epithelial cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2012; 1819:869-76. [DOI: 10.1016/j.bbagrm.2012.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 04/13/2012] [Accepted: 04/17/2012] [Indexed: 02/07/2023]
|
27
|
GATA6 is required for proliferation, migration, secretory cell maturation, and gene expression in the mature mouse colon. Mol Cell Biol 2012; 32:3392-402. [PMID: 22733991 DOI: 10.1128/mcb.00070-12] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Controlled renewal of the epithelium with precise cell distribution and gene expression patterns is essential for colonic function. GATA6 is expressed in the colonic epithelium, but its function in the colon is currently unknown. To define GATA6 function in the colon, we conditionally deleted Gata6 throughout the epithelium of small and large intestines of adult mice. In the colon, Gata6 deletion resulted in shorter, wider crypts, a decrease in proliferation, and a delayed crypt-to-surface epithelial migration rate. Staining techniques and electron microscopy indicated deficient maturation of goblet cells, and coimmunofluorescence demonstrated alterations in specific hormones produced by the endocrine L cells and serotonin-producing cells. Specific colonocyte genes were significantly downregulated. In LS174T, the colonic adenocarcinoma cell line, Gata6 knockdown resulted in a significant downregulation of a similar subset of goblet cell and colonocyte genes, and GATA6 was found to occupy active loci in enhancers and promoters of some of these genes, suggesting that they are direct targets of GATA6. These data demonstrate that GATA6 is necessary for proliferation, migration, lineage maturation, and gene expression in the mature colonic epithelium.
Collapse
|
28
|
Skrypek N, Duchêne B, Hebbar M, Leteurtre E, van Seuningen I, Jonckheere N. The MUC4 mucin mediates gemcitabine resistance of human pancreatic cancer cells via the Concentrative Nucleoside Transporter family. Oncogene 2012; 32:1714-23. [PMID: 22580602 DOI: 10.1038/onc.2012.179] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The fluorinated analog of deoxycytidine, Gemcitabine (Gemzar), is the main chemotherapeutic drug in pancreatic cancer, but survival remains weak mainly because of the high resistance of tumors to the drug. Recent works have shown that the mucin MUC4 may confer an advantage to pancreatic tumor cells by modifying their susceptibility to drugs. However, the cellular mechanism(s) responsible for this MUC4-mediated resistance is unknown. The aim of this work was to identify the cellular mechanisms responsible for gemcitabine resistance linked to MUC4 expression. CAPAN-2 and CAPAN-1 adenocarcinomatous pancreatic cancer (PC) cell lines were used to establish stable MUC4-deficient clones (MUC4-KD) by shRNA interference. Measurement of the IC50 index using tetrazolium salt test indicated that MUC4-deficient cells were more sensitive to gemcitabine. This was correlated with increased Bax/BclXL ratio and apoptotic cell number. Expression of Equilibrative/Concentrative Nucleoside Transporter (hENT1, hCNT1/3), deoxycytidine kinase (dCK), ribonucleotide reductase (RRM1/2) and Multidrug-Resistance Protein (MRP3/4/5) was evaluated by quantitative RT-PCR (qRT-PCR) and western blotting. Alteration of MRP3, MRP4, hCNT1 and hCNT3 expression was observed in MUC4-KD cells, but only hCNT1 alteration was correlated to MUC4 expression and sensitivity to gemcitabine. Decreased activation of MAPK, JNK and NF-κB pathways was observed in MUC4-deficient cells, in which the NF-κB pathway was found to have an important role in both sensitivity to gemcitabine and hCNT1 regulation. Finally, and in accordance with our in vitro data, we found that MUC4 expression was conversely correlated to that of hCNT1 in tissues from patients with pancreatic adenocarcinoma. This work describes a new mechanism of PC cell resistance to gemcitabine, in which the MUC4 mucin negatively regulates the hCNT1 transporter expression via the NF-κB pathway. Altogether, these data point out to MUC4 and hCNT1 as potential targets to ameliorate the response of pancreatic tumors to gemcitabine treatment.
Collapse
Affiliation(s)
- N Skrypek
- Inserm, UMR837, Jean-Pierre Aubert Research Center, Lille Cedex, France
| | | | | | | | | | | |
Collapse
|
29
|
Jonckheere N, Skrypek N, Merlin J, Dessein AF, Dumont P, Leteurtre E, Harris A, Desseyn JL, Susini C, Frénois F, Van Seuningen I. The mucin MUC4 and its membrane partner ErbB2 regulate biological properties of human CAPAN-2 pancreatic cancer cells via different signalling pathways. PLoS One 2012; 7:e32232. [PMID: 22393391 PMCID: PMC3290552 DOI: 10.1371/journal.pone.0032232] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 01/24/2012] [Indexed: 01/19/2023] Open
Abstract
The mucin MUC4 and its membrane partner the ErbB2 oncogenic receptor are potential interacting partners in human pancreatic tumour development. However, the way they function is still largely unknown. In this work, we aimed to identify the cellular mechanisms and the intracellular signalling pathways under the control of both ErbB2 and MUC4 in a human pancreatic adenocarcinomatous cell line. Using co-immunoprecipitation and GST pull-down, we show that MUC4 and ErbB2 interact in the human pancreatic adenocarcinomatous cell line CAPAN-2 via the EGF domains of MUC4. Stable cell clones were generated in which either MUC4 or ErbB2 were knocked down (KD) by a shRNA approach. Biological properties of these cells were then studied in vitro and in vivo. Our results show that ErbB2-KD cells are more apoptotic and less proliferative (decreased cyclin D1 and increased p27kip1 expression) while migration and invasive properties were not altered. MUC4-KD clones were less proliferative with decreased cyclin D1 expression, G1 cell cycle arrest and altered ErbB2/ErbB3 expression. Their migration properties were reduced whereas invasive properties were increased. Importantly, inhibition of ErbB2 and MUC4 expression did not impair the same signalling pathways (inhibition of MUC4 expression affected the JNK pathway whereas that of ErbB2 altered the MAPK pathway). Finally, ErbB2-KD and MUC4-KD cells showed impaired tumour growth in vivo. Our results show that ErbB2 and MUC4, which interact physically, activate different intracellular signalling pathways to regulate biological properties of CAPAN-2 pancreatic cancer cells.
Collapse
Affiliation(s)
- Nicolas Jonckheere
- Inserm, UMR837, Jean Pierre Aubert Research Center, Team #5 Mucins, epithelial differentiation and carcinogenesis, Lille, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Wouthuyzen-Bakker M, Bijvelds MJ, de Jonge HR, De Lisle RC, Burgerhof JG, Verkade HJ. Effect of antibiotic treatment on fat absorption in mice with cystic fibrosis. Pediatr Res 2012; 71:4-12. [PMID: 22289844 PMCID: PMC3786567 DOI: 10.1038/pr.2011.4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Improving fat absorption remains a challenge in cystic fibrosis (CF). Antibiotics (AB) treatment has been shown to improve body weight in CF mice. The mechanism may include improvement in fat absorption. We aimed to determine the effect of AB on fat absorption in two CF mouse models. RESULTS AB did not improve total fat absorption. Interestingly, AB accelerated the absorption of isotope-labeled fats, in both Δ/Δ and WT mice. The changes observed were not related to the solubilization capacity of bile or to changes in the bacteria in the small intestine. AB reduced the fecal excretion of cholate by ~50% (P < 0.05) in both CF mouse models, indicating improved intestinal bile salt absorption. DISCUSSION In conclusion, AB treatment does not improve total fat absorption in CF mice but does decrease fecal loss of bile salts and accelerate long-chain fatty acid (LCFA) absorption. METHODS For 3 weeks, we administered oral AB (ciprofloxacin/metronidazole) or control treatment to homozygous ΔF508 (Δ/Δ), cystic fibrosis transmembrane conductance regulator (CFTR) knockout (-/-), and wild-type (WT) mice and quantified fat absorption using a 72-h fat balance test. In Δ/Δ mice, we assessed fat absorption kinetics by administering tri-1-(13)C-palmitin and 1-(13)C-stearate intragastrically and determining the appearance of stable isotope-labeled fats in plasma. We quantified biliary and fecal bile salts (gas chromatography) and small intestinal bacteria (quantitative-PCR).
Collapse
Affiliation(s)
- Marjan Wouthuyzen-Bakker
- Department of Pediatrics, Beatrix Children's Hospital–University Medical Center Groningen, Groningen, The Netherlands,Department of Pediatric Gastroenterology, Beatrix Children's Hospital–University Medical Center Groningen, Groningen, The Netherlands
| | - Marcel J.C. Bijvelds
- Laboratory of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hugo R. de Jonge
- Laboratory of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Robert C. De Lisle
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, Missouri
| | - Johannes G.M. Burgerhof
- Department of Epidemiology, Unit Medical Statistics, University Medical Center Groningen, Groningen, The Netherlands
| | - Henkjan J. Verkade
- Department of Pediatrics, Beatrix Children's Hospital–University Medical Center Groningen, Groningen, The Netherlands,Department of Pediatric Gastroenterology, Beatrix Children's Hospital–University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
31
|
Bai H, Sakurai T, Konno T, Ideta A, Aoyagi Y, Godkin JD, Imakawa K. Expression of GATA1 in the ovine conceptus and endometrium during the peri-attachment period. Mol Reprod Dev 2011; 79:64-73. [PMID: 22102538 DOI: 10.1002/mrd.21409] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 10/26/2011] [Indexed: 11/10/2022]
Abstract
The transcription factor GATA1 is known to play an essential role in hematopoiesis, but its other roles have not been well characterized. The purpose of this study was to determine relationships between GATA1 and GATA2 and/or GATA3, and to identify their possible functions in ovine development. GATA1 mRNA was found in ovine conceptuses and endometrial epithelial regions of Day 15 (Day 0=day of estrus) cyclic and Days 15, 17, and 21 pregnant ovine uteri. GATA1 mRNA was strongly expressed in conceptuses on Day 21, when trophoblast attachment to the maternal endometrium progressed. Similarly, GATA1 protein expression was relatively high on Day 21. To localize GATA1 mRNA, ovine conceptuses and pregnant uteri were subjected to in situ hybridization on Days 15, 17, and 21, confirming that GATA1 mRNA was expressed in trophoblasts and uterine endometrial epithelial cells in these gestation days. The presence of GATA1 protein was further confirmed by immunohistochemistry. Because high GATA1 expression appeared to coincide with reduced GATA2/3 expression, a potential role of GATA1 was examined through transfection of a mouse Gata1 expression plasmid into bovine trophoblast F3 cells. This over-expression resulted in the down-regulation of endogenous GATA2 transcripts. These observations indicate that GATA1 exists in the ovine conceptus and uterus during the peri-attachment period, and suggest that GATA1 is integral to conceptus and endometrial development through the regulation of GATA2 and possibly other developmentally important genes.
Collapse
Affiliation(s)
- Hanako Bai
- Laboratory of Animal Breeding, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
32
|
Lu P, Burger-van Paassen N, van der Sluis M, Witte-Bouma J, Kerckaert JP, van Goudoever JB, Van Seuningen I, Renes IB. Colonic gene expression patterns of mucin Muc2 knockout mice reveal various phases in colitis development. Inflamm Bowel Dis 2011; 17:2047-57. [PMID: 21910166 DOI: 10.1002/ibd.21592] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 11/02/2010] [Indexed: 12/26/2022]
Abstract
BACKGROUND Mucin Muc2 knockout (Muc2(-/-)) mice spontaneously develop colitis. METHODS To identify genes and biological responses which play a pivotal role during colitis development in Muc2(-/-) mice, gene expression profiles of colonic tissues from 2- and 4-week-old Muc2(-/-) and wildtype mice were determined using microarrays. RESULTS The majority of highly upregulated genes in 2-week-old as well as 4-week-old Muc2(-/-) mice were primarily involved in immune responses related to antigen processing/presentation, B-cell and T-cell receptor signaling, leukocyte transendothelial migration, and Jak-STAT signaling. Specifically, Muc2(-/-) mice expressed high levels of immunoglobulins, murine histocompatibility-2, proinflammatory cytokines, chemokines, and antimicrobial proteins. Additionally, in 4-week-old Muc2(-/-) mice, expression of genes involved in cell structure related pathways was significantly altered. Particularly, the tight junction-associated gene claudin-10 was upregulated, whereas claudin-1 and claudin-5 were downregulated. Furthermore, 4-week-old Muc2(-/-) mice showed increased expression of genes regulating cell growth in conjunction with increased crypt length and increased epithelial proliferation. CONCLUSIONS Muc2-deficiency leads to an active inflammatory response in 2- and 4-week-old Muc2(-/-) mice as demonstrated by the altered expression in immune response related genes. In addition, 4-week-old Muc2(-/-) mice also showed a decrease in epithelial barrier function and an increase in epithelial proliferation as indicated by, respectively, the altered expression in tight junction-related genes and upregulation of genes stimulating cell growth. Remarkably, upregulation of genes stimulating cell growth correlated with increased crypt length and increased epithelial proliferation in 4-week-old Muc2(-/-) mice. Together, these data demonstrate that there are distinct phases in colitis development in 2-4-week-old Muc2(-/-) mice.
Collapse
Affiliation(s)
- Peng Lu
- Laboratory of Pediatrics, Division Neonatology, Erasmus MC-Sophia, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Bruyère E, Jonckheere N, Frénois F, Mariette C, Van Seuningen I. The MUC4 membrane-bound mucin regulates esophageal cancer cell proliferation and migration properties: Implication for S100A4 protein. Biochem Biophys Res Commun 2011; 413:325-9. [DOI: 10.1016/j.bbrc.2011.08.095] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 08/19/2011] [Indexed: 12/24/2022]
|
34
|
|
35
|
Jonckheere N, Velghe A, Ducourouble MP, Copin MC, Renes IB, Van Seuningen I. The mouse Muc5b mucin gene is transcriptionally regulated by thyroid transcription factor-1 (TTF-1) and GATA-6 transcription factors. FEBS J 2010; 278:282-94. [PMID: 21126317 DOI: 10.1111/j.1742-4658.2010.07945.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
MUC5B is one of the major mucin genes expressed in the respiratory tract. Previous studies in our laboratory have demonstrated that MUC5B is expressed in human lung adenocarcinomas and during lung morphogenesis. Moreover, in human lung adenocarcinoma tissues, a converse correlation between MUC5B and thyroid transcription factor-1 (TTF-1) expression, a lung-specific transcription factor, has been established. However, the molecular mechanisms that govern the regulation of MUC5B expression in the lung are largely unknown. In order to better understand the biological role of MUC5B in lung pathophysiology, we report the characterization of the promoter region of the mouse Muc5b mucin gene. The promoter is flanked by a TATA box (TACATAA) identical to that in the human gene. Human and murine promoters share 67.5% similarity over the first 170 nucleotides. By RT-PCR, co-transfection studies and gel-shift assays, we show that Muc5b promoter activity is completely inhibited by TTF-1, whereas factors of the GATA family (GATA-4/GATA-5/GATA-6) are activators. Together, these results demonstrate, for the first time, that Muc5b is a target gene of transcription factors (TTF-1, GATA-6) involved in lung differentiation programs during development and carcinogenesis, and identify TTF-1 as a strong repressor of Muc5b. The characterization of the structural and functional features of the Muc5b mucin gene will provide us with a strong base to develop studies in murine models aimed at the identification of its biological role in lung pathophysiology.
Collapse
|
36
|
Jonckheere N, Skrypek N, Van Seuningen I. Mucins and pancreatic cancer. Cancers (Basel) 2010; 2:1794-812. [PMID: 24281201 PMCID: PMC3840449 DOI: 10.3390/cancers2041794] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 10/14/2010] [Accepted: 10/18/2010] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is characterized by an often dramatic outcome (five year survival < 5%) related to a late diagnosis and a lack of efficient therapy. Therefore, clinicians desperately need new biomarkers and new therapeutic tools to develop new efficient therapies. Mucins belong to an ever increasing family of O-glycoproteins. Secreted mucins are the main component of mucus protecting the epithelia whereas membrane-bound mucins are thought to play important biological roles in cell-cell and cell-matrix interactions, in cell signaling and in modulating biological properties of cancer cells. In this review, we will focus on the altered expression pattern of mucins in pancreatic cancer, from the early neoplastic lesion Pancreatic Intraepithelial Neoplasia (PanIN) to invasive pancreatic carcinomas, and the molecular mechanisms (including genetic and epigenetic regulation) and signaling pathways known to control their expression. Moreover, we will discuss the recent advances about the biology of both secreted and membrane-bound mucins and their key roles in pancreatic carcinogenesis and resistance to therapy. Finally, we will discuss exciting opportunities that mucins offer as potential therapeutic targets in pancreatic cancer.
Collapse
Affiliation(s)
- Nicolas Jonckheere
- INSERM, U837, Jean-Pierre Aubert Research Center, Team 5 "Mucins, epithelial differentiation and carcinogenesis", Lille, France.
| | | | | |
Collapse
|
37
|
Rapp UR, Korn C, Ceteci F, Karreman C, Luetkenhaus K, Serafin V, Zanucco E, Castro I, Potapenko T. MYC is a metastasis gene for non-small-cell lung cancer. PLoS One 2009; 4:e6029. [PMID: 19551151 PMCID: PMC2696940 DOI: 10.1371/journal.pone.0006029] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Accepted: 05/25/2009] [Indexed: 11/19/2022] Open
Abstract
Background Metastasis is a process by which cancer cells learn to form satellite tumors in distant organs and represents the principle cause of death of patients with solid tumors. NSCLC is the most lethal human cancer due to its high rate of metastasis. Methodology/Principal Findings Lack of a suitable animal model has so far hampered analysis of metastatic progression. We have examined c-MYC for its ability to induce metastasis in a C-RAF-driven mouse model for non-small-cell lung cancer. c-MYC alone induced frank tumor growth only after long latency at which time secondary mutations in K-Ras or LKB1 were detected reminiscent of human NSCLC. Combination with C-RAF led to immediate acceleration of tumor growth, conversion to papillary epithelial cells and angiogenic switch induction. Moreover, addition of c-MYC was sufficient to induce macrometastasis in liver and lymph nodes with short latency associated with lineage switch events. Thus we have generated the first conditional model for metastasis of NSCLC and identified a gene, c-MYC that is able to orchestrate all steps of this process. Conclusions/Significance Potential markers for detection of metastasis were identified and validated for diagnosis of human biopsies. These markers may represent targets for future therapeutic intervention as they include genes such as Gata4 that are exclusively expressed during lung development.
Collapse
Affiliation(s)
- Ulf R Rapp
- Department of Molecular Biology, Max-Planck-Institute of Biochemistry, München, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Yamaguchi K, Cekanova M, McEntee MF, Yoon JH, Fischer SM, Renes IB, Van Seuningen I, Baek SJ. Peroxisome proliferator-activated receptor ligand MCC-555 suppresses intestinal polyps in ApcMin/+ mice via extracellular signal-regulated kinase and peroxisome proliferator-activated receptor-dependent pathways. Mol Cancer Ther 2008; 7:2779-87. [PMID: 18790758 PMCID: PMC2597004 DOI: 10.1158/1535-7163.mct-08-0173] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
A large body of studies has suggested that peroxisome proliferator-activated receptor gamma (PPARgamma) ligands, such as thiazolidinedione, are potent candidates for chemopreventive agents. MCC-555 is a PPARgamma/alpha dual agonist and has been shown previously to induce apoptosis in vitro; however, the molecular mechanisms by which MCC-555 affects antitumorigenesis in vivo are poorly understood. In this study, we explored the antitumorigenic effects of MCC-555 both in cell culture and in Apc-deficient mice, an animal model for human familial adenomatous polyposis. MCC-555 increased MUC2 expression in colorectal and lung cancer cells, and treatment with the PPARgamma antagonist GW9662 revealed that MUC2 induction by MCC-555 was mediated in a PPARgamma-dependent manner. Moreover, MCC-555 increased transcriptional activity of human and mouse MUC2 promoters. Subsequently, treatment with MCC-555 (30 mg/kg/d) for 4 weeks reduced the number of small intestinal polyps to 54.8% of that in control mice. In agreement with in vitro studies, enhanced Muc2 expression was observed in the small intestinal tumors of Min mice treated with MCC-555, suggesting that MUC2 expression may be associated at least in part with the antitumorigenic action of MCC-555. In addition, highly phosphorylated extracellular signal-regulated kinase (ERK) was found in the intestinal tumors of MCC-555-treated Min mice, and inhibition of the ERK pathway by a specific inhibitor markedly suppressed MCC-555-induced Muc2 expression in vitro. Overall, these results indicate that MCC-555 has a potent tumor suppressor activity in intestinal tumorigenesis, likely involving MUC2 up-regulation by ERK and PPARgamma pathways.
Collapse
Affiliation(s)
- Kiyoshi Yamaguchi
- Department of Pathobiology, College of Veterinary Medicine, The University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
van der Sluis M, Vincent A, Bouma J, Male AKV, van Goudoever JB, Renes IB, Van Seuningen I. Forkhead box transcription factors Foxa1 and Foxa2 are important regulators of Muc2 mucin expression in intestinal epithelial cells. Biochem Biophys Res Commun 2008; 369:1108-13. [DOI: 10.1016/j.bbrc.2008.02.158] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Accepted: 02/28/2008] [Indexed: 10/22/2022]
|
40
|
Haveri H, Westerholm-Ormio M, Lindfors K, Mäki M, Savilahti E, Andersson LC, Heikinheimo M. Transcription factors GATA-4 and GATA-6 in normal and neoplastic human gastrointestinal mucosa. BMC Gastroenterol 2008; 8:9. [PMID: 18405344 PMCID: PMC2323380 DOI: 10.1186/1471-230x-8-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2007] [Accepted: 04/11/2008] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Human gastrointestinal mucosa regenerates vigorously throughout life, but the factors controlling cell fate in mature mucosa are poorly understood. GATA transcription factors direct cell proliferation and differentiation in many organs, and are implicated in tumorigenesis. GATA-4 and GATA-6 are considered crucial for the formation of murine gastrointestinal mucosa, but their role in human gastrointestinal tract remains unexplored. We studied in detail the expression patterns of these two GATA factors and a GATA-6 down-stream target, Indian hedgehog (Ihh), in normal human gastrointestinal mucosa. Since these factors are considered important for proliferation and differentiation, we also explored the possible alterations in their expression in gastrointestinal neoplasias. The expression of the carcinogenesis-related protein Indian hedgehog was also investigated in comparison to GATA factors. METHODS Samples of normal and neoplastic gastrointestinal tract from children and adults were subjected to RNA in situ hybridization with 33P labelled probes and immunohistochemistry, using an avidin-biotin immunoperoxidase system. The pathological tissues examined included samples of chronic and atrophic gastritis as well as adenomas and adenocarcinomas of the colon and rectum. RESULTS GATA-4 was abundant in the differentiated epithelial cells of the proximal parts of the gastrointestinal tract but was absent from the distal parts. In contrast, GATA-6 was expressed throughout the gastrointestinal epithelium, and in the distal gut its expression was most intense at the bottom of the crypts, i.e. cells with proliferative capacity. Both factors were also present in Barrett's esophagus and metaplasia of the stomach. GATA-6 expression was reduced in colon carcinoma. Ihh expression overlapped with that of GATA-6 especially in benign gastrointestinal neoplasias. CONCLUSION The results suggest differential but overlapping functions for GATA-4 and GATA-6 in the normal gastrointestinal mucosa. Furthermore, GATA-4, GATA-6 and Ihh expression is altered in premalignant dysplastic lesions and reduced in overt cancer.
Collapse
Affiliation(s)
- Hanna Haveri
- Children's Hospital, University of Helsinki, Helsinki, Finland.
| | | | | | | | | | | | | |
Collapse
|
41
|
Linden SK, Sutton P, Karlsson NG, Korolik V, McGuckin MA. Mucins in the mucosal barrier to infection. Mucosal Immunol 2008; 1:183-97. [PMID: 19079178 PMCID: PMC7100821 DOI: 10.1038/mi.2008.5] [Citation(s) in RCA: 860] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The mucosal tissues of the gastrointestinal, respiratory, reproductive, and urinary tracts, and the surface of the eye present an enormous surface area to the exterior environment. All of these tissues are covered with resident microbial flora, which vary considerably in composition and complexity. Mucosal tissues represent the site of infection or route of access for the majority of viruses, bacteria, yeast, protozoa, and multicellular parasites that cause human disease. Mucin glycoproteins are secreted in large quantities by mucosal epithelia, and cell surface mucins are a prominent feature of the apical glycocalyx of all mucosal epithelia. In this review, we highlight the central role played by mucins in accommodating the resident commensal flora and limiting infectious disease, interplay between underlying innate and adaptive immunity and mucins, and the strategies used by successful mucosal pathogens to subvert or avoid the mucin barrier, with a particular focus on bacteria.
Collapse
Affiliation(s)
- S K Linden
- grid.1003.20000 0000 9320 7537Mucosal Diseases Program, Mater Medical Research Institute and The University of Queensland, Level 3 Aubigny Place, Mater Hospitals, South Brisbane, Queensland Australia
| | - P Sutton
- grid.1008.90000 0001 2179 088XCentre for Animal Biotechnology, School of Veterinary Science, University of Melbourne, Melbourne, Victoria Australia
| | - N G Karlsson
- grid.6142.10000 0004 0488 0789Department of Chemistry, Centre for BioAnalytical Sciences, National University of Ireland, Galway, Ireland
| | - V Korolik
- grid.1022.10000 0004 0437 5432Institute for Glycomics, Griffith University, Gold Coast, Queensland Australia
| | - M A McGuckin
- grid.1003.20000 0000 9320 7537Mucosal Diseases Program, Mater Medical Research Institute and The University of Queensland, Level 3 Aubigny Place, Mater Hospitals, South Brisbane, Queensland Australia
| |
Collapse
|
42
|
Fauquette V, Aubert S, Groux-Degroote S, Hemon B, Porchet N, Van Seuningen I, Pigny P. Transcription factor AP-2alpha represses both the mucin MUC4 expression and pancreatic cancer cell proliferation. Carcinogenesis 2007; 28:2305-12. [PMID: 17621592 DOI: 10.1093/carcin/bgm158] [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] [Indexed: 11/13/2022] Open
Abstract
MUC4 is a transmembrane mucin expressed in pancreatic ductal adenocarcinoma (DAC) in contrast to normal pancreas, and is an independent predictor of poor prognosis in patients with invasive DAC. Our aim was therefore to investigate the mechanisms that control MUC4 expression in pancreatic cancer cells. We focused our study on activator protein (AP)-2alpha transcription factor that acts as a tumour suppressor gene in several cancers. In a series of 18 human DAC, using immunohistochemistry, we confirmed that MUC4 was exclusively expressed in cancerous or preneoplastic lesions in 83% of the samples. On the contrary, AP-2 was mainly expressed by non-tumoural ductal cells (61%) or endocrine cells (67%). Moreover, MUC4 and AP-2 were never found co-expressed suggesting an inhibitory role of AP-2alpha in normal ductal cells. In CAPAN-1 and CAPAN-2 cells, transient AP-2alpha over-expression decreased both MUC4 mRNA and apomucin levels by 20-40% by a mechanism involving inhibition of MUC4 promoter. By chromatin immunoprecipitation and gel-shift assays, we demonstrated that this inhibition involved two AP-2 cis-elements located in the -475/-238 region of the promoter. CAPAN-1 clones, which stably over-expressed AP-2alpha, displayed a strong MUC4 down-regulation (-38 to -100%), a significant decrease of both cell proliferation and invasion concomitant to the up-regulation of p27 cyclin-dependent kinase inhibitor. In conclusion, our data provide evidence that AP-2alpha is an important in vivo negative regulator of MUC4 expression in human pancreatic tissue and that AP-2alpha may play a tumour-suppressive role in pancreatic DAC.
Collapse
|
43
|
Jonckheere N, Vincent A, Perrais M, Ducourouble MP, Male AKV, Aubert JP, Pigny P, Carraway KL, Freund JN, Renes IB, Van Seuningen I. The human mucin MUC4 is transcriptionally regulated by caudal-related homeobox, hepatocyte nuclear factors, forkhead box A, and GATA endodermal transcription factors in epithelial cancer cells. J Biol Chem 2007; 282:22638-50. [PMID: 17553805 DOI: 10.1074/jbc.m700905200] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The human gene MUC4 encodes a large transmembrane mucin that is developmentally regulated and expressed along the undifferentiated pseudostratified epithelium, as early as 6.5 weeks during fetal development. Immunohistochemical analysis of Muc4 expression in developing mouse lung and gastrointestinal tract showed a different spatio-temporal pattern of expression before and after cytodifferentiation. The molecular mechanisms governing MUC4 expression during development are, however, unknown. Hepatocyte nuclear factors (HNF), forkhead box A (FOXA), GATA, and caudal-related homeobox transcription factors (TFs) are known to control cell differentiation of gut endoderm derived-tissues during embryonic development. They also control the expression of cell- and tissue-specific genes and may thus control MUC4 expression. To test this hypothesis, we studied and deciphered the molecular mechanisms responsible for MUC4 transcriptional regulation by these TFs. Experiments using small interfering RNA, cell co-transfection, and site-directed mutagenesis indicated that MUC4 is regulated at the transcriptional level by CDX-1 and -2, HNF-1 alpha and -1 beta, FOXA1/A2, HNF-4 alpha and -4 gamma, and GATA-4, -5, and -6 factors in a cell-specific manner. Binding of TFs was assessed by chromatin immunoprecipitation, and gel-shift assays. Altogether, these results demonstrate that MUC4 is a target gene of endodermal TFs and thus point out an important role for these TFs in regulating MUC4 expression during epithelial differentiation during development, cancer, and repair.
Collapse
|
44
|
Piessen G, Jonckheere N, Vincent A, Hémon B, Ducourouble MP, Copin MC, Mariette C, Seuningen I. Regulation of the human mucin MUC4 by taurodeoxycholic and taurochenodeoxycholic bile acids in oesophageal cancer cells is mediated by hepatocyte nuclear factor 1alpha. Biochem J 2007; 402:81-91. [PMID: 17037983 PMCID: PMC1783985 DOI: 10.1042/bj20061461] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
MUC4 (mucin 4) is a membrane-bound mucin overexpressed in the early steps of oesophageal carcinogenesis and implicated in tumour progression. We previously showed that bile acids, main components of gastro-oesophageal reflux and tumour promoters, up-regulate MUC4 expression [Mariette, Perrais, Leteurtre, Jonckheere, Hemon, Pigny, Batra, Aubert, Triboulet and Van Seuningen (2004) Biochem. J. 377, 701-708]. HNF (hepatocyte nuclear factor) 1alpha and HNF4alpha transcription factors are known to mediate bile acid effects, and we previously identified cis-elements for these factors in MUC4 distal promoter. Our aim was to demonstrate that these two transcription factors were directly involved in MUC4 activation by bile acids. MUC4, HNF1alpha and HNF4alpha expressions were evaluated by immunohistochemistry in human oesophageal tissues. Our results indicate that MUC4, HNF1alpha and HNF4alpha were co-expressed in oesophageal metaplastic and adenocarcinomatous tissues. Studies at the mRNA, promoter and protein levels indicated that HNF1alpha regulates endogenous MUC4 expression by binding to two cognate cis-elements respectively located at -3332/-3327 and -3040/-3028 in the distal promoter. We also showed by siRNA (small interfering RNA) approach, co-transfection and site-directed mutagenesis that HNF1alpha mediates taurodeoxycholic and taurochenodeoxycholic bile acid activation of endogenous MUC4 expression and transcription in a dose-dependent manner. In conclusion, these results describe a new mechanism of regulation of MUC4 expression by bile acids, in which HNF1alpha is a key mediator. These results bring new insights into MUC4 up-regulation in oesophageal carcinoma associated with bile reflux.
Collapse
Affiliation(s)
- Guillaume Piessen
- *Unité INSERM 560, Place de Verdun, 59045 Lille Cedex, France
- †Department of Digestive and Oncological Surgery, C. Huriez Hospital, Centre Hospitalier Régional et Universitaire de Lille, 59037 Lille Cedex, France
| | | | - Audrey Vincent
- *Unité INSERM 560, Place de Verdun, 59045 Lille Cedex, France
| | - Brigitte Hémon
- *Unité INSERM 560, Place de Verdun, 59045 Lille Cedex, France
| | | | - Marie-Christine Copin
- *Unité INSERM 560, Place de Verdun, 59045 Lille Cedex, France
- ‡Department of Pathology, Parc Eurasanté, CHRU Lille, 59037 Lille Cedex, France
| | - Christophe Mariette
- *Unité INSERM 560, Place de Verdun, 59045 Lille Cedex, France
- †Department of Digestive and Oncological Surgery, C. Huriez Hospital, Centre Hospitalier Régional et Universitaire de Lille, 59037 Lille Cedex, France
| | - Isabelle VAN Seuningen
- *Unité INSERM 560, Place de Verdun, 59045 Lille Cedex, France
- To whom correspondence should be addressed (email )
| |
Collapse
|
45
|
Van der Sluis M, De Koning BAE, De Bruijn ACJM, Velcich A, Meijerink JPP, Van Goudoever JB, Büller HA, Dekker J, Van Seuningen I, Renes IB, Einerhand AWC. Muc2-deficient mice spontaneously develop colitis, indicating that MUC2 is critical for colonic protection. Gastroenterology 2006; 131:117-29. [PMID: 16831596 DOI: 10.1053/j.gastro.2006.04.020] [Citation(s) in RCA: 1205] [Impact Index Per Article: 63.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2005] [Accepted: 03/23/2006] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Expression of mucin MUC2, the structural component of the colonic mucus layer, is lowered in inflammatory bowel disease. Our aim was to obtain insight in the role of Muc2 in epithelial protection. METHODS Muc2 knockout (Muc2(-/-)) and Muc2 heterozygous (Muc2(+/-)) mice were characterized and challenged by a colitis-inducing agent, dextran sulfate sodium (DSS). We monitored clinical symptoms, intestinal morphology, and differences in intestine-specific protein and messenger RNA levels. RESULTS The Muc2(-/-) mice showed clinical signs of colitis (as of 5 weeks), aggravating as the mice aged. Microscopic analysis of the colon of Muc2(-/-) mice showed mucosal thickening, increased proliferation, and superficial erosions. Colonic goblet cells in the Muc2(-/-) mice were negative for Muc2, but trefoil factor 3 was still detectable. In Muc2(-/-) mice, transient de novo expression of Muc6 messenger RNA was observed in the distal colon. On day 2 of DSS treatment, the histologic damage was more severe in Muc2(+/-) versus wild-type (Muc2(+/+)) mice, but the disease activity index was not yet different. By day 7, the disease activity index and histologic score were significantly elevated in Muc2(+/-) versus Muc2(+/+) mice. The disease activity index of the Muc2(-/-) mice was higher (versus both Muc2(+/+) and Muc2(+/-) mice) throughout DSS treatment. The histologic damage in the DSS-treated Muc2(-/-) mice was different compared with Muc2(+/+) and Muc2(+/-) mice, with many crypt abscesses instead of mucosal ulcerations. CONCLUSIONS This study shows that Muc2 deficiency leads to inflammation of the colon and contributes to the onset and perpetuation of experimental colitis.
Collapse
Affiliation(s)
- Maria Van der Sluis
- Division of Neonatology, Department of Pediatrics, Erasmus MC and Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abba MC, Nunez MI, Colussi AG, Croce MV, Segal-Eiras A, Aldaz CM. GATA3 protein as a MUC1 transcriptional regulator in breast cancer cells. Breast Cancer Res 2006; 8:R64. [PMID: 17078870 PMCID: PMC1797033 DOI: 10.1186/bcr1617] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2006] [Revised: 07/22/2006] [Accepted: 11/01/2006] [Indexed: 01/24/2023] Open
Abstract
INTRODUCTION Recent studies have demonstrated that members of the GATA-binding protein (GATA) family (GATA4 and GATA5) might have pivotal roles in the transcriptional upregulation of mucin genes (MUC2, MUC3 and MUC4) in gastrointestinal epithelium. The zinc-finger GATA3 transcription factor has been reported to be involved in the growth control and differentiation of breast epithelial cells. In SAGE (serial analysis of gene expression) studies we observed an intriguing significant correlation between GATA3 and MUC1 mRNA expression in breast carcinomas. We therefore designed the present study to elucidate whether MUC1 expression is regulated by GATA3 in breast cancer cells. METHODS Promoter sequence analysis of the MUC1 gene identified six GATA cis consensus elements in the 5' flanking region (GATA1, GATA3 and four GATA-like sequences). Chromatin immunoprecipitation and electrophoretic mobility-shift assays were employed to study the presence of a functional GATA3-binding site. GATA3 and MUC1 expression was analyzed in vitro with a GATA3 knockdown assay. Furthermore, expression of GATA3 and MUC1 genes was analyzed by real-time RT-PCR and immunohistochemistry on breast cancer-specific tissue microarrays. RESULTS We confirmed the presence of a functional GATA3-binding site on the MUC1 promoter region in the MCF7 cell line. We determined that GATA3 knockdown assays led to a decrease in MUC1 protein expression in MCF7 and T47D cells. In addition, we detected a statistically significant correlation in expression between GATA3 and MUC1 genes at the mRNA and protein levels both in normal breast epithelium and in breast carcinomas (p = 0.01). GATA3 expression was also highly associated with estrogen receptor and progesterone receptor status (p = 0.0001) and tumor grade (p = 0.004) in breast carcinomas. CONCLUSION Our study provides evidence indicating that GATA3 is probably a mediator for the transcriptional upregulation of MUC1 expression in some breast cancers.
Collapse
Affiliation(s)
- Martín C Abba
- Department of Carcinogenesis, The University of Texas MD Anderson Cancer Center, Science Park – Research Division, PO Box 389, Smithville, TX 78957, USA
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas (CINIBA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, CP:1900, Argentina
| | - María I Nunez
- Department of Carcinogenesis, The University of Texas MD Anderson Cancer Center, Science Park – Research Division, PO Box 389, Smithville, TX 78957, USA
| | - Andrea G Colussi
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas (CINIBA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, CP:1900, Argentina
| | - María V Croce
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas (CINIBA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, CP:1900, Argentina
| | - Amada Segal-Eiras
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas (CINIBA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, CP:1900, Argentina
| | - C Marcelo Aldaz
- Department of Carcinogenesis, The University of Texas MD Anderson Cancer Center, Science Park – Research Division, PO Box 389, Smithville, TX 78957, USA
| |
Collapse
|