Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 286(21): 18515-25, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21388956

ABSTRACT

Fibroblast growth factor receptor-3 (FGFR-3) expression in the developing intestine is restricted to the undifferentiated epithelial cells within the lower portion of the crypt. We previously showed that mice lacking functional FGFR-3 have a significant decrease in the number of Paneth cells in the small intestine. Here, we used Caco2 cells to investigate whether FGFR-3 signaling can directly modulate expression of Paneth cell differentiation markers through its effects on TCF4/ß-catenin or through other signaling pathways downstream of this receptor. Caco2 cells treated with FGFR-3 ligands or expressing FGFR-3(K650E), a constitutively active mutant, resulted in a significantly increased expression of genes characteristic of mature Paneth cells, including human α-defensins 5 and 6 (HD5 and HD6) and Paneth cell lysozyme, whereas enterocytic differentiation markers were reduced. Activation of FGFR-3 signaling sustained high levels of ß-catenin mRNA expression, leading to increased TCF4/ß-catenin-regulated transcriptional activity in Caco2 cells. Sustained activity of the TCF4/ß-catenin pathway was required for the induction of Paneth cell markers. Activation of the MAPK pathway by FGFR-3 is also required for the induction of Paneth cell markers in addition to and independent of the effect of FGFR-3 on TCF4/ß-catenin activity. These studies suggest that coordinate activation of multiple independent signaling pathways downstream of FGFR-3 is involved in regulation of Paneth cell differentiation.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation/physiology , MAP Kinase Signaling System/physiology , Paneth Cells/metabolism , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Transcription Factors/metabolism , beta Catenin/metabolism , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Caco-2 Cells , Cell Differentiation/physiology , Humans , Lysosomes/genetics , Lysosomes/metabolism , Mice , Mutation, Missense , Organ Specificity/physiology , Receptor, Fibroblast Growth Factor, Type 3/genetics , Transcription Factor 4 , Transcription Factors/genetics , alpha-Defensins/genetics , alpha-Defensins/metabolism , beta Catenin/genetics
2.
J Biol Chem ; 285(10): 7493-504, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20056603

ABSTRACT

Paneth cells at the base of small intestinal crypts of Lieberkühn secrete host defense peptides and proteins, including alpha-defensins, as mediators of innate immunity. Mouse Paneth cells also express alpha-defensin-related Defcr-rs genes that code for cysteine-rich sequence 4C (CRS4C) peptides that have a unique CPX triplet repeat motif. In ileitis-prone SAMP1/YitFc mice, Paneth cell levels of CRS4C mRNAs and peptides are induced more than a 1000-fold relative to non-prone strains as early as 4 weeks of age, with the mRNA and peptide levels highest in distal ileum and below detection in duodenum. CRS4C-1 peptides are found exclusively in Paneth cells where they occur only in dense core granules and thus are secreted to function in the intestinal lumen. CRS4C bactericidal peptide activity is membrane-disruptive in that it permeabilizes Escherichia coli and induces rapid microbial cell K(+) efflux, but in a manner different from mouse alpha-defensin cryptdin-4. In in vitro studies, inactive pro-CRS4C-1 is converted to bactericidal CRS4C-1 peptide by matrix metalloproteinase-7 (MMP-7) proteolysis of the precursor proregion at the same residue positions that MMP-7 activates mouse pro-alpha-defensins. The absence of processed CRS4C in protein extracts of MMP-7-null mouse ileum demonstrates the in vivo requirement for intracellular MMP-7 in pro-CRS4C processing.


Subject(s)
Defensins/metabolism , Ileitis/metabolism , Paneth Cells/metabolism , Protein Precursors/metabolism , Animals , Anti-Bacterial Agents/metabolism , Defensins/genetics , Ileum/cytology , Ileum/metabolism , Ileum/pathology , Matrix Metalloproteinase 7/genetics , Matrix Metalloproteinase 7/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Protein Precursors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Secretory Vesicles/metabolism , Secretory Vesicles/ultrastructure , Tissue Distribution , alpha-Defensins/genetics , alpha-Defensins/metabolism
3.
Int J Parasitol ; 40(7): 833-43, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20083116

ABSTRACT

Entamoeba histolytica contains a large and novel family of transmembrane kinases (TMKs). The expression patterns of the E. histolytica TMKs in individual trophozoites and the roles of the TMKs for sensing and responding to extracellular cues were incompletely characterised. Here we provide evidence that single cells express multiple TMKs and that TMK39 and TMK54 likely serve non-redundant cellular functions. Laser-capture microdissection was used in conjunction with microarray analysis to demonstrate that single trophozoites express more than one TMK gene. Anti-peptide antibodies were raised against unique regions in the extracellular domains of TMK39, TMK54 and PaTMK, and TMK expression was analysed at the protein level. Flow cytometric assays revealed that populations of trophozoites homogeneously expressed TMK39, TMK54 and PaTMK, while confocal microscopy identified different patterns of cell surface expression for TMK39 and TMK54. The functions of TMK39 and TMK54 were probed by the inducible expression of dominant-negative mutants. While TMK39 co-localised with ingested beads and expression of truncated TMK39 interfered with trophozoite phagocytosis of apoptotic lymphocytes, expression of a truncated TMK54 inhibited growth of amoebae and altered the surface expression of the heavy subunit of the E. histolytica Gal/GalNAc lectin. Overall, our data indicates that multiple members of the novel E. histolytica TMK family are utilised for non-redundant functions by the parasite.


Subject(s)
Entamoeba histolytica/physiology , Phagocytosis , Protozoan Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Entamoeba histolytica/growth & development , Flow Cytometry , Gene Expression Profiling , Microscopy, Confocal , Microscopy, Fluorescence , Oligonucleotide Array Sequence Analysis
4.
Am J Physiol Gastrointest Liver Physiol ; 297(4): G632-40, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19696144

ABSTRACT

Intestinal cell kinase (ICK), originally cloned from the intestine and expressed in the intestinal crypt epithelium, is a highly conserved serine/threonine protein kinase that is similar to mitogen-activated protein kinases (MAPKs) in the catalytic domain and requires dual phosphorylation within a MAPK-like TDY motif for full activation. Despite these similarities to MAPKs, the biological functions of ICK remain unknown. In this study, we report that suppression of ICK expression in cultured intestinal epithelial cells by short hairpin RNA (shRNA) interference significantly impaired cellular proliferation and induced features of gene expression characteristic of colonic or enterocytic differentiation. Downregulation of ICK altered expression of cell cycle regulators (cyclin D1, c-Myc, and p21(Cip1/WAF1)) of G(1)-S transition, consistent with the G(1) cell cycle delay induced by ICK shRNA. ICK deficiency also led to a significant decrease in the expression and/or activity of p70 ribosomal protein S6 kinase (S6K1) and eukaryotic initiation factor 4E (eIF4E), concomitant with reduced expression of their upstream regulators, the mammalian target of rapamycin (mTOR) and the regulatory associated protein of mTOR (Raptor). Furthermore, ICK interacts with the mTOR/Raptor complex in vivo and phosphorylates Raptor in vitro. These results suggest that disrupting ICK function may downregulate protein translation of specific downstream targets of eIF4E and S6K1 such as cyclin D1 and c-Myc through the mTOR/Raptor signaling pathway. Taken together, our findings demonstrate an important role for ICK in proliferation and differentiation of intestinal epithelial cells.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation , Epithelial Cells/enzymology , G1 Phase , Intestinal Mucosa/enzymology , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Caco-2 Cells , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epithelial Cells/pathology , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation , Humans , Intestinal Mucosa/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proteins , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Regulatory-Associated Protein of mTOR , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Transcription Factors/metabolism , Transfection
5.
Am J Physiol Gastrointest Liver Physiol ; 297(1): G168-78, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19407216

ABSTRACT

Fibroblast growth factor receptor 3 (FGFR-3) is expressed in the lower crypt epithelium, where stem cells of the intestine reside. The role of FGFR-3 signaling in regulating features of intestinal morphogenesis was examined in FGFR-3-null (FGFR-3(-/-)) mice. FGFR-3(-/-) mice had only about half the number of intestinal crypts and a marked decrease in the number of functional clonogenic stem cells, as assessed by an in vivo microcolony-forming assay, compared with wild-type littermates. A marked deficit in allocation of progenitor cells to Paneth cell differentiation was noted, although all the principal epithelial lineages were represented in FGFR-3(-/-) mice. The total cellular content and nuclear localization of beta-catenin protein were reduced in FGFR-3(-/-) mice, as was expression of cyclin D1 and matrix metalloproteinase-7, major downstream targets of beta-catenin/T cell factor-4 (Tcf-4) signaling. Activation of FGFR-3 in Caco-2 cells, an intestinal epithelial cell line, abrogated the fall in beta-catenin/Tcf-4 signaling activity that is normally observed in these cells as cultures become progressively more confluent. These findings are consistent with the hypothesis that, during intestinal development, FGFR-3 signaling regulates crypt epithelial stem cell expansion and crypt morphogenesis, as well as Paneth cell lineage specification, through beta-catenin/Tcf-4-dependent and -independent pathways.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Movement , Cell Proliferation , Intestinal Mucosa/metabolism , Paneth Cells/metabolism , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Stem Cells/metabolism , Age Factors , Aging/metabolism , Animals , Caco-2 Cells , Cyclin D1/metabolism , Humans , Intestines/growth & development , Matrix Metalloproteinase 7/metabolism , Mice , Mice, Knockout , Phenotype , Receptor, Fibroblast Growth Factor, Type 3/deficiency , Receptor, Fibroblast Growth Factor, Type 3/genetics , Signal Transduction , TCF Transcription Factors/metabolism , Transcription Factor 7-Like 2 Protein , beta Catenin/metabolism
6.
J Immunol ; 179(10): 7012-20, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17982092

ABSTRACT

SAMP1/Fc mice develop spontaneous ileitis that shares many features with human Crohn's disease. One of the earliest features of ileitis in SAMP1/Fc mice is an increase in the number of ileal goblet and intermediate cells. Resistin-like molecule beta (RELMbeta) is a goblet cell-specific, cysteine-rich peptide previously shown to function as part of the innate immune response. In this study, we examined the role of expression of RELMbeta in the initiation of ileal inflammation in SAMP1/Fc mice. RELMbeta was highly induced in the ilea of SAMP1/Fc mice beginning at age 5 wk, coincident with the histological appearance of inflammation. RELMbeta was found in ileal goblet cells and some intermediate and Paneth cells. Surprisingly, RELMbeta mRNA levels were significantly increased in the ilea of 80% of germ-free SAMP1/Fc mice examined compared with specific pathogen-free AKR control mice of similar age. Ileitis was observed in germfree SAMP1/Fc mice, although it was attenuated relative to specific pathogen-free SAMP1/Fc mice. These data suggest that neither the early induction of RELMbeta expression nor ileal inflammation requires the presence of viable intestinal flora. Neither was the induction of RELMbeta dependent on the major Th1 or Th2 cytokines. However, RELMbeta stimulated naive bone marrow-derived macrophages to secrete significant amounts of TNF-alpha, IL-6, and RANTES. Our data suggest that RELMbeta is involved in the initiation of ileitis in SAMP1/Fc mice and may act through the induction of proinflammatory cytokines from resident immune cells within the mucosa.


Subject(s)
Goblet Cells/immunology , Hormones, Ectopic/immunology , Ileitis/immunology , Paneth Cells/immunology , Animals , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/pathology , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Gene Expression Regulation/immunology , Goblet Cells/pathology , Hormones, Ectopic/genetics , Hormones, Ectopic/pharmacology , Ileitis/genetics , Ileitis/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators/immunology , Intercellular Signaling Peptides and Proteins , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Transgenic , Paneth Cells/pathology , Th1 Cells/immunology , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology
7.
Am J Pathol ; 166(4): 1055-67, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15793286

ABSTRACT

Crohn's disease is characterized by cycles of mucosal injury and ulceration followed by epithelial regeneration and restoration of normal epithelial function. In this study, we examined whether ileitis in SAMP1/YitFc mice, a recombinant-inbred line that spontaneously develops ileitis resembling human Crohn's disease, was associated with alterations in normal patterns of epithelial differentiation or changes in epithelial regeneration after experimental injury. Increased numbers of Paneth, goblet, and intermediate cells were present focally in the ileum of SAMP1/YitFc mice by 4 weeks of age, before any histological evidence of acute or chronic inflammation. This increase in secretory cells became more pronounced at sites of ileitis with increasing age and inflammation. Additionally, there was mispositioning of Paneth and intermediate cells along the crypt-to-villus unit. A concomitant reduction in the number of absorptive enterocytes was observed. In contrast to the ileal-specific changes in lineage allocation, crypt stem cell numbers began to increase in both the ileum and proximal jejunum at the onset of inflammation in SAMP1/YitFc mice. These data suggest that the alterations in epithelial cell differentiation and increases in the size of the crypt stem cell population observed in SAMP1/YitFc mice are regulated by distinct mechanisms. We speculate that these epithelial alterations may play a role in the pathogenesis of ileitis in this murine model of Crohn's disease.


Subject(s)
Epithelial Cells/cytology , Ileitis/pathology , Intestinal Mucosa/cytology , Stem Cells/cytology , Animals , Apoptosis , Cell Differentiation/physiology , Cell Differentiation/radiation effects , Cell Lineage , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Ileitis/metabolism , Immunohistochemistry , Inflammation/metabolism , Inflammation/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/radiation effects , Mice , Mice, Mutant Strains , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Stem Cells/radiation effects
8.
Dev Dyn ; 230(1): 114-23, 2004 May.
Article in English | MEDLINE | ID: mdl-15108315

ABSTRACT

Prior studies have demonstrated that fibroblast growth factor receptor-3 (FGFR-3) regulates proliferation of undifferentiated intestinal epithelial cells in vitro. However, the function(s) of FGFR-3-mediated signaling during intestinal development and epithelial differentiation in vivo remain unknown. The goal of this study was to define the temporal, regional, and cell-specific patterns of FGFR-3 expression and its ligands during normal intestinal ontogeny and epithelial regeneration. Both the IIIb and IIIc isoforms of FGFR-3 mRNA, which result from differential splicing of the FGFR-3 primary transcript, were detected in mouse small intestine as early as embryonic day 16. FGFR-3 levels peaked in the small intestine from 7 to 21 days after birth and decreased thereafter to reach the low levels observed in adult mice. FGFR-3 IIIb and IIIc mRNA levels were highest in the duodenum and proximal jejunum with lower levels of both seen in the distal jejunum, ileum, and colon. FGFR-3 was expressed in a subset of proliferating undifferentiated crypt epithelial cells located in the intervillous epithelium and in the lower half of nascently forming crypts but not in differentiated epithelial cell types. FGFR-3 IIIb was the dominant isoform expressed in both small intestinal and colonic crypts. Expression of FGF1, FGF2, and FGF9, known ligands of FGFR-3, paralleled patterns of FGFR-3 expression during gut development. These data suggest that signaling through FGFR-3 plays a role in regulating morphogenic events involved in formation of intestinal crypts and/or the fate of epithelial stem cells.


Subject(s)
Gene Expression Regulation, Developmental , Intestinal Mucosa/metabolism , Intestines/embryology , Protein-Tyrosine Kinases/biosynthesis , Receptors, Fibroblast Growth Factor/biosynthesis , Animals , Bromodeoxyuridine/pharmacology , Cell Differentiation , Cell Division , Epithelium/embryology , Immunohistochemistry , Intestine, Small/embryology , Ligands , Mice , Morphogenesis , Protein Isoforms , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 3 , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tissue Distribution
9.
Gastroenterology ; 124(4): 972-82, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12671894

ABSTRACT

BACKGROUND & AIMS: SAMP1/Yit mice spontaneously develop chronic terminal ileitis, reminiscent of the human disease described by Crohn et al. in 1932. Several new phenotypic features have appeared in our colony after more than 20 generations of brother-sister mating. In this report, we describe the distinguishing features of the SAMP1/YitFc substrain at the University of Virginia, compared with the Japanese SAMP1/Yit parental strain. METHODS: A colony of SAMP1/Yit mice was established at the University of Virginia in 1996, from 2 breeding pairs obtained from Japan. A systematic characterization of their phenotypic and immunologic characteristics was performed at 4, 10, 40, and more than 60 weeks of age. RESULTS: The following differences were observed: (1) SAMP1/YitFc mice displayed established ileitis as early as 10 weeks of age, (2) the incidence of skin lesions inversely correlated with the occurrence of intestinal inflammation, (3) mice develop chronic ileitis with prominent muscular hypertrophy and focal collagen deposition in inflamed segments, (4) mesenteric lymph node lymphocytes acquired an activated phenotype coincident with disease progression, (5) high interferon-gamma production was detected by 4 weeks of age and preceded the onset of ileitis, and (6) a subgroup of mice (approximately 5%) developed perianal disease with ulceration and fistulae. CONCLUSIONS: The SAMP1/YitFc substrain exhibits unique characteristics when compared with the original Japanese strain. Of particular interest is the emergence of perianal fistulizing disease, to our knowledge the first report of such occurrence in an animal model of inflammatory bowel disease.


Subject(s)
Ileitis/genetics , Ileitis/pathology , Perianal Glands/pathology , Rectal Fistula/genetics , Rectal Fistula/pathology , Animals , Cells, Cultured , Chronic Disease , Cytokines/metabolism , Disease Models, Animal , Female , Flow Cytometry , Hypertrophy , Ileum/pathology , Lymph Nodes/cytology , Lymph Nodes/metabolism , Male , Mesenteric Lymphadenitis/genetics , Mesenteric Lymphadenitis/pathology , Mice , Mice, Mutant Strains , Muscle, Smooth/pathology , Phenotype , Pregnancy , Skin/pathology , Specific Pathogen-Free Organisms
10.
Curr Opin Gastroenterol ; 19(6): 583-90, 2003 Nov.
Article in English | MEDLINE | ID: mdl-15703609

ABSTRACT

PURPOSE OF REVIEW: In the past year, the study of intestinal stem cell biology has realized significant progress toward understanding the mechanisms and pathways regulating crypt stem cell turnover, maintenance, and differentiation. RECENT FINDINGS: This review summarizes recent investigations that have contributed significantly to the elucidation of mechanisms operative during intestinal development and in the adult intestine that regulate maintenance of the stem cell niche, cell fate and lineage allocation, and establishment and maintenance of the architectural organization of the crypt-to-villus axis. SUMMARY: The relevance of the findings discussed in this review extends beyond the field of intestinal development to encompass the study of tissue remodeling and repair and intestinal neoplasia.

SELECTION OF CITATIONS
SEARCH DETAIL
...