Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 11(520)2019 11 27.
Article in English | MEDLINE | ID: mdl-31776290

ABSTRACT

Cerebral cavernous malformation (CCM) is a genetic, cerebrovascular disease. Familial CCM is caused by genetic mutations in KRIT1, CCM2, or PDCD10 Disease onset is earlier and more severe in individuals with PDCD10 mutations. Recent studies have shown that lesions arise from excess mitogen-activated protein kinase kinase kinase 3 (MEKK3) signaling downstream of Toll-like receptor 4 (TLR4) stimulation by lipopolysaccharide derived from the gut microbiome. These findings suggest a gut-brain CCM disease axis but fail to define it or explain the poor prognosis of patients with PDCD10 mutations. Here, we demonstrate that the gut barrier is a primary determinant of CCM disease course, independent of microbiome configuration, that explains the increased severity of CCM disease associated with PDCD10 deficiency. Chemical disruption of the gut barrier with dextran sulfate sodium augments CCM formation in a mouse model, as does genetic loss of Pdcd10, but not Krit1, in gut epithelial cells. Loss of gut epithelial Pdcd10 results in disruption of the colonic mucosal barrier. Accordingly, loss of Mucin-2 or exposure to dietary emulsifiers that reduce the mucus barrier increases CCM burden analogous to loss of Pdcd10 in the gut epithelium. Last, we show that treatment with dexamethasone potently inhibits CCM formation in mice because of the combined effect of action at both brain endothelial cells and gut epithelial cells. These studies define a gut-brain disease axis in an experimental model of CCM in which a single gene is required for two critical components: gut epithelial function and brain endothelial signaling.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Brain/metabolism , Gastrointestinal Tract/metabolism , Hemangioma, Cavernous, Central Nervous System/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Brain/pathology , Carrier Proteins/metabolism , Colitis/complications , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Dextran Sulfate , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/pathology , Hemangioma, Cavernous, Central Nervous System/drug therapy , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , KRIT1 Protein/metabolism , Ligands , Mice , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism
2.
Oncotarget ; 8(42): 71456-71470, 2017 Sep 22.
Article in English | MEDLINE | ID: mdl-29069719

ABSTRACT

The mucus layer in the intestine affects several aspects of intestinal biology, encompassing physical, chemical protection, immunomodulation and growth, thus contributing to homeostasis. Mice with genetic inactivation of the Muc2 gene, encoding the MUC2 mucin, the major protein component of mucus, exhibit altered intestinal homeostasis, which is strictly dependent on the habitat, likely due to differing complements of intestinal microbes. Our previous work established that Muc2 deficiency was linked to low chronic inflammation resulting in tumor development in the small, large intestine including the rectum. Here, we report that inactivation of Muc2 alters metabolic pathways in the normal appearing mucosa of Muc2-/- mice. Comparative analysis of gene expression profiling of isolated intestinal epithelial cells (IECs) and the entire intestinal mucosa, encompassing IECs, immune and stromal cells underscored that more than 50% of the changes were common to both sets of data, suggesting that most alterations were IEC-specific. IEC-specific expression data highlighted perturbation of lipid absorption, processing and catabolism linked to altered Pparα signaling in IECs. Concomitantly, alterations of glucose metabolism induced expression of genes linked to de novo lipogenesis, a characteristic of tumor cells. Importantly, gene expression alterations characterizing Muc2-/- IECs are similar to those observed when analyzing the gene expression signature of IECs along the crypt-villus axis in WT B6 mice, suggesting that Muc2-/- IECs display a crypt-like gene expression signature. Thus, our data strongly suggest that decreased lipid metabolism, and alterations in glucose utilization characterize the crypt proliferative compartment, and may represent a molecular signature of pre-neoplastic lesions.

3.
Dose Response ; 15(1): 1559325816685798, 2017.
Article in English | MEDLINE | ID: mdl-28203121

ABSTRACT

BACKGROUND AND PURPOSE: Currently, no readily available mitigators exist for acute abdominal radiation injury. Here, we present an animal model for precise and homogenous limb-sparing abdominal irradiation (LSAIR) to study the radiation-induced gastrointestinal syndrome (RIGS). MATERIALS AND METHODS: The LSAIR technique was developed using the small animal radiation research platform (SARRP) with image guidance capabilities. We delivered LSAIR at doses between 14 and 18 Gy on 8- to 10-week-old male C57BL/6 mice. Histological analysis was performed to confirm that the observed mortality was due to acute abdominal radiation injury. RESULTS: A steep dose-response relationship was found for survival, with no deaths seen at doses below 16 Gy and 100% mortality at above 17 Gy. All deaths occurred between 6 and 10 days after irradiation, consistent with the onset of RIGS. This was further confirmed by histological analysis showing clear differences in the number of regenerative intestinal crypts between animals receiving sublethal (14 Gy) and 100% lethal (18 Gy) radiation. CONCLUSION: The developed LSAIR technique provides uniform dose delivery with a clear dose response, consistent with acute abdominal radiation injury on histological examination. This model can provide a useful tool for researchers investigating the development of mitigators for accidental or clinical high-dose abdominal irradiation.

4.
J Immunol ; 194(2): 606-14, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25480561

ABSTRACT

Expression of a germline VH3609/D/JH2 IgH in mice results in the generation of B1 B cells with anti-thymocyte/Thy-1 glycoprotein autoreactivity by coexpression of Vk21-5/Jk2 L chain leading to production of serum IgM natural autoantibody. In these same mice, the marginal zone (MZ) B cell subset in spleen shows biased usage of a set of Ig L chains different from B1 B cells, with 30% having an identical Vk19-17/Jk1 L chain rearrangement. This VH3609/Vk19-17 IgM is reactive with intestinal goblet cell granules, binding to the intact large polymatrix form of mucin 2 glycoprotein secreted by goblet cells. Analysis of a µκ B cell AgR (BCR) transgenic (Tg) mouse with this anti-goblet cell/mucin2 autoreactive (AGcA) specificity demonstrates that immature B cells expressing the Tg BCR become MZ B cells in spleen by T cell-independent BCR signaling. These Tg B cells produce AGcA as the predominant serum IgM, but without enteropathy. Without the transgene, AGcA autoreactivity is low but detectable in the serum of BALB/c and C.B17 mice, and this autoantibody is specifically produced by the MZ B cell subset. Thus, our findings reveal that AGcA is a natural autoantibody associated with MZ B cells.


Subject(s)
Autoantibodies/immunology , B-Lymphocyte Subsets/immunology , Goblet Cells/immunology , Mucin-2/immunology , Receptors, Antigen, B-Cell/immunology , Secretory Vesicles/immunology , Animals , Autoantibodies/genetics , B-Lymphocyte Subsets/pathology , Goblet Cells/pathology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin M/genetics , Immunoglobulin M/immunology , Immunoglobulin kappa-Chains/genetics , Immunoglobulin kappa-Chains/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mucin-2/genetics , Receptors, Antigen, B-Cell/genetics , Secretory Vesicles/genetics , Secretory Vesicles/pathology , Spleen/immunology , Spleen/pathology
5.
PLoS One ; 9(6): e100217, 2014.
Article in English | MEDLINE | ID: mdl-24945909

ABSTRACT

BACKGROUND: The colonic mucus layer plays a critical role in intestinal homeostasis by limiting contact between luminal bacteria and the mucosal immune system. A defective mucus barrier in animal models allows bacterial contact with the intestinal epithelium and results in spontaneous colitis. A defective mucus barrier is also a key feature of active ulcerative colitis (UC). Alterations in the immune compartment due to intestinal bacterial breach in mice lacking the colon mucus barrier have not been characterized and correlated to active UC. AIMS: To characterize alterations in the immune compartment due to intestinal bacterial breach in Muc2-/- mice, which lack the colon mucus barrier, and correlate the findings to active UC. METHODS: Bacterial contact with colon epithelium and penetration into colon tissue was examined in Muc2-/- mice and colon biopsies from patients with active UC using fluorescence microscopy and qPCR. Neutrophils, lymphocytes, CD103+ dendritic cell subsets and macrophages in colon from Muc2-/- mice and biopsies from UC patients were quantitated by flow cytometry. RESULTS: Inflamed UC patients and Muc2-/- mice had bacteria in contact with the colon epithelium. Bacterial rRNA was present in colonic mucosa in humans and Muc2-/- mice and in the draining lymph nodes of mice. Inflamed Muc2-/- mice and UC patients had elevated colon neutrophils, T cells and macrophages while a reduced frequency of CD103+ DCs was present in the inflamed colon of both mice and humans. CONCLUSIONS: The parallel features of the colon immune cell compartment in Muc2-/- mice and UC patients supports the usefulness of this model to understand the early phase of spontaneous colitis and will provide insight into novel strategies to treat UC.


Subject(s)
Colitis, Ulcerative/pathology , Mucin-2/deficiency , Adult , Aged , Animals , Antigens, CD/metabolism , Cell Count , Colitis, Ulcerative/microbiology , Colon/microbiology , Colon/pathology , Dendritic Cells/pathology , Female , Humans , Inflammation/pathology , Integrin alpha Chains/metabolism , Intestinal Mucosa/pathology , Lipopolysaccharide Receptors/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mucin-2/metabolism , Neutrophil Infiltration , Young Adult
6.
Cancer Biother Radiopharm ; 29(4): 153-61, 2014 May.
Article in English | MEDLINE | ID: mdl-24693958

ABSTRACT

We sought to determine if single-dose external beam radiation therapy (EBRT) could modulate the expression signature of T-cell costimulatory and coinhibitory molecules in human prostate cancer (PCa) cell lines in vitro. We investigated the functional impact of irradiated PCa cells with a modulated costimulatory profile on responder T-cell activity. We used three PCa cell lines (DU145, PC3, and LNCaP) and two epithelial cell lines from noncancerous prostate and lung tissue. After 72 hours of EBRT, surface expression of four immunostimulatory molecules (CD70, CD275/ICOSL, CD134L/OX40L, and CD137L/41BBL) and two immunosuppressive markers (CTLA-4/CD152 and PD-L1/CD274) were evaluated by flow cytometry. We evaluated the impact of several radiation doses and the longevity of modulated expression. We examined the functional impact of radiation-induced modulation of cancer cells by cytotoxic T cells (CTL) cytotoxicity and ELISPOT assay for interferon-gamma (IFN-γ) production. Last, we evaluated whether IFN-γ-induced PD-L1 expression could be reversed by EBRT. After 10 Gy EBRT, expression of OX40L and 41BBL increased in all three PCa cell lines; expression of CD70 and ICOSL increased in PC3 cells. Conversely, a decrease in PD-L1 expression in DU145 and PC3 cells was detectable up to 144 hours after EBRT. No PD-L1 was detected in LNCaP. Epithelial cells from normal prostate were not modulated by radiation. CTL cytolytic activity and IFN-γ production were enhanced by interaction with irradiated PCa cells. Finally, EBRT failed to prevent IFN-γ-induced upregulation of PD-L1. We demonstrate that a single dose of EBRT increased surface expression of costimulatory molecules and decreased the expression of coinhibitory molecules in human PCa cell lines. Changes in irradiated tumor cells led to functional enhancement of T-cell activity, despite EBRT failing to reduce IFN-γ-induced expression of PD-L1. These data suggest that combining radiotherapy with T-cell stimulating immunotherapy may be an attractive strategy for cancer treatment.


Subject(s)
Costimulatory and Inhibitory T-Cell Receptors/immunology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/radiotherapy , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/radiation effects , Animals , Cell Line, Tumor , Humans , Male , Mice, Inbred C57BL , Prostatic Neoplasms/pathology , Signal Transduction
7.
Science ; 342(6157): 447-53, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24072822

ABSTRACT

A dense mucus layer in the large intestine prevents inflammation by shielding the underlying epithelium from luminal bacteria and food antigens. This mucus barrier is organized around the hyperglycosylated mucin MUC2. Here we show that the small intestine has a porous mucus layer, which permitted the uptake of MUC2 by antigen-sampling dendritic cells (DCs). Glycans associated with MUC2 imprinted DCs with anti-inflammatory properties by assembling a galectin-3-Dectin-1-FcγRIIB receptor complex that activated ß-catenin. This transcription factor interfered with DC expression of inflammatory but not tolerogenic cytokines by inhibiting gene transcription through nuclear factor κB. MUC2 induced additional conditioning signals in intestinal epithelial cells. Thus, mucus does not merely form a nonspecific physical barrier, but also constrains the immunogenicity of gut antigens by delivering tolerogenic signals.


Subject(s)
Homeostasis , Immune Tolerance/immunology , Intestine, Small/immunology , Mouth/immunology , Mucus/immunology , Animals , Cells, Cultured , Dendritic Cells/immunology , Galectin 3/genetics , Galectin 3/metabolism , Glycosylation , Humans , Immune Tolerance/genetics , Inflammation/immunology , Intestinal Mucosa/immunology , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mucin-2/genetics , Mucin-2/physiology , NF-kappa B/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Transcription, Genetic , beta Catenin/metabolism
8.
Infect Immun ; 81(10): 3672-83, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23876803

ABSTRACT

Salmonella enterica serovar Typhimurium is a model organism used to explore the virulence strategies underlying Salmonella pathogenesis. Although intestinal mucus is the first line of defense in the intestine, its role in protection against Salmonella is still unclear. The intestinal mucus layer is composed primarily of the Muc2 mucin, a heavily O-glycosylated glycoprotein. The core 3-derived O-glycans of Muc2 are synthesized by core 3 ß1,3-N-acetylglucosaminyltransferase (C3GnT). Mice lacking these glycans still produce Muc2 but display a thinner intestinal mucus barrier. We began our investigations by comparing Salmonella-induced colitis and mucus dynamics in Muc2-deficient (Muc2(-/-)) mice, C3GnT(-/-) mice, and wild-type C57BL/6 (WT) mice. Salmonella infection led to increases in luminal Muc2 secretion in WT and C3GnT(-/-) mice. When Muc2(-/-) mice were infected with Salmonella, they showed dramatic susceptibility to infection, carrying significantly higher cecal and liver pathogen burdens, and developing significantly higher barrier disruption and higher mortality rates, than WT mice. We found that the exaggerated barrier disruption in infected Muc2(-/-) mice was invA dependent. We also tested the susceptibility of C3GnT(-/-) mice and found that they carried pathogen burdens similar to those of WT mice but developed exaggerated barrier disruption. Moreover, we found that Muc2(-/-) mice were impaired in intestinal alkaline phosphatase (IAP) expression and lipopolysaccharide (LPS) detoxification activity in their ceca, potentially explaining their high mortality rates during infection. Our data suggest that the intestinal mucus layer (Muc2) and core 3 O-glycosylation play critical roles in controlling Salmonella intestinal burdens and intestinal epithelial barrier function, respectively.


Subject(s)
Colitis/microbiology , Gene Expression Regulation/physiology , Intestinal Mucosa/pathology , Mucin-2/metabolism , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/physiology , Animals , Colitis/pathology , Intestinal Mucosa/microbiology , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-2/genetics , Polysaccharides , Salmonella Infections, Animal/pathology
9.
PLoS Pathog ; 6(5): e1000902, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20485566

ABSTRACT

Despite recent advances in our understanding of the pathogenesis of attaching and effacing (A/E) Escherichia coli infections, the mechanisms by which the host defends against these microbes are unclear. The goal of this study was to determine the role of goblet cell-derived Muc2, the major intestinal secretory mucin and primary component of the mucus layer, in host protection against A/E pathogens. To assess the role of Muc2 during A/E bacterial infections, we inoculated Muc2 deficient (Muc2(-/-)) mice with Citrobacter rodentium, a murine A/E pathogen related to diarrheagenic A/E E. coli. Unlike wildtype (WT) mice, infected Muc2(-/-) mice exhibited rapid weight loss and suffered up to 90% mortality. Stool plating demonstrated 10-100 fold greater C. rodentium burdens in Muc2(-/-) vs. WT mice, most of which were found to be loosely adherent to the colonic mucosa. Histology of Muc2(-/-) mice revealed ulceration in the colon amid focal bacterial microcolonies. Metabolic labeling of secreted mucins in the large intestine demonstrated that mucin secretion was markedly increased in WT mice during infection compared to uninfected controls, suggesting that the host uses increased mucin release to flush pathogens from the mucosal surface. Muc2 also impacted host-commensal interactions during infection, as FISH analysis revealed C. rodentium microcolonies contained numerous commensal microbes, which was not observed in WT mice. Orally administered FITC-Dextran and FISH staining showed significantly worsened intestinal barrier disruption in Muc2(-/-) vs. WT mice, with overt pathogen and commensal translocation into the Muc2(-/-) colonic mucosa. Interestingly, commensal depletion enhanced C. rodentium colonization of Muc2(-/-) mice, although colonic pathology was not significantly altered. In conclusion, Muc2 production is critical for host protection during A/E bacterial infections, by limiting overall pathogen and commensal numbers associated with the colonic mucosal surface. Such actions limit tissue damage and translocation of pathogenic and commensal bacteria across the epithelium.


Subject(s)
Citrobacter rodentium , Colitis/immunology , Enterobacteriaceae Infections/immunology , Intestinal Mucosa/immunology , Mucin-2/metabolism , Animals , Bacterial Adhesion/immunology , Bacterial Translocation/immunology , Colitis/metabolism , Colitis/microbiology , Enterobacteriaceae Infections/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mucin-2/genetics , Mucin-2/immunology
10.
Gastroenterology ; 138(5): 1763-71, 2010 May.
Article in English | MEDLINE | ID: mdl-20138044

ABSTRACT

BACKGROUND & AIMS: Hyperplasia of mucin-secreting intestinal goblet cells accompanies a number of enteric infections, including infections by nematode parasites. Nevertheless, the precise role of mucins in host defense in nematode infection is not known. We investigated the role of the mucin (Muc2) in worm expulsion and host immunity in a model of nematode infection. METHODS: Resistant (BALB/c, C57BL/6), susceptible (AKR), and Muc2-deficient mouse strains were infected with the nematode, Trichuris muris, and worm expulsion, energy status of the whipworms, changes in mucus/mucins, and inflammatory and immune responses were investigated after infection. RESULTS: The increase in Muc2 production, observed exclusively in resistant mice, correlated with worm expulsion. Moreover, expulsion of the worms from the intestine was significantly delayed in the Muc2-deficient mice. Although a marked impairment in the development of periodic acid Schiff (PAS)-stained intestinal goblet cells was observed in Muc2-deficient mice, as infection progressed a significant increase in the number of PAS-positive goblet cells was observed in these mice. Surprisingly, an increase in Muc5ac, a mucin normally expressed in the airways and stomach, was observed after infection of only the resistant animals. Overall, the mucus barrier in the resistant mice was less permeable than that of susceptible mice. Furthermore, the worms isolated from the resistant mice had a lower energy status. CONCLUSIONS: Mucins are an important component of innate defense in enteric infection; this is the first demonstration of the important functional contribution of mucins to host protection from nematode infection.


Subject(s)
Goblet Cells/metabolism , Intestinal Diseases, Parasitic/metabolism , Mucin-2/deficiency , Trichuriasis/metabolism , Trichuris/pathogenicity , Adenosine Triphosphate/metabolism , Animals , Disease Models, Animal , Energy Metabolism , Goblet Cells/immunology , Goblet Cells/parasitology , Immunity, Innate , Immunity, Mucosal , Intestinal Diseases, Parasitic/genetics , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/parasitology , Intestinal Diseases, Parasitic/prevention & control , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Mucin 5AC/metabolism , Mucin-2/genetics , Permeability , Species Specificity , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/parasitology , Time Factors , Trichuriasis/genetics , Trichuriasis/immunology , Trichuriasis/parasitology , Trichuriasis/prevention & control , Trichuris/immunology , Trichuris/metabolism
11.
J Pediatr Gastroenterol Nutr ; 49(1): 99-107, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19502996

ABSTRACT

OBJECTIVES: Previous studies have shown that the intestine uses a major part of the dietary threonine intake for the synthesis of the structural component of the protective intestinal mucus layer, the secretory mucin Muc2. In this context, the high intestinal demand for dietary threonine probably results from its incorporation into secretory mucins rich in threonine residues. Therefore, we compared threonine utilization in the colon of Muc2 knockout (Muc2-/-) and wild-type (Muc2+/+) mice to investigate the intestinal dietary threonine metabolism in the absence of Muc2, which results in inflammation of the colon. MATERIALS AND METHODS: Concentrations and isotopic enrichment of threonine were measured by gas chromatography-isotope ratio mass spectrometry in the serum, colon, and colonic content of mice given a bolus [U-(13)C]threonine enterally. RESULTS: We retrieved 37.8% and 40.9% of dietary threonine in Muc2 +/+ and Muc2 -/- mice, respectively, either as free or incorporated threonine. There were no major differences in the availability and concentration of free or incorporated threonine recovered in both serum and colon in both types of mice. However, the Muc2 -/- mice did show overall significantly higher threonine oxidation rates compared with Muc2 +/+ mice. CONCLUSIONS: In the absence of Muc2, dietary threonine is mainly used for constitutive protein synthesis or becomes a substrate for metabolic oxidation. This indicates that inflammation also requires high threonine amounts.


Subject(s)
Intestine, Large/metabolism , Mucin-2/metabolism , Threonine/metabolism , Animals , Female , Intestinal Mucosa/metabolism , Isotopes , Mass Spectrometry , Mice , Mice, Knockout , Mucin-2/genetics , Threonine/genetics
12.
Exp Cell Res ; 315(8): 1415-28, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19232344

ABSTRACT

Alterations in PKC isozyme expression and aberrant induction of cyclin D1 are early events in intestinal tumorigenesis. Previous studies have identified cyclin D1 as a major target in the antiproliferative effects of PKCalpha in non-transformed intestinal cells; however, a link between PKC signaling and cyclin D1 in colon cancer remained to be established. The current study further characterized PKC isozyme expression in intestinal neoplasms and explored the consequences of restoring PKCalpha or PKCdelta in a panel of colon carcinoma cell lines. Consistent with patterns of PKC expression in primary tumors, PKCalpha and delta levels were generally reduced in colon carcinoma cell lines, PKCbetaII was elevated and PKCepsilon showed variable expression, thus establishing the suitability of these models for analysis of PKC signaling. While colon cancer cells were insensitive to the effects of PKC agonists on cyclin D1 levels, restoration of PKCalpha downregulated cyclin D1 by two independent mechanisms. PKCalpha expression consistently (a) reduced steady-state levels of cyclin D1 by a novel transcriptional mechanism not previously seen in non-transformed cells, and (b) re-established the ability of PKC agonists to activate the translational repressor 4E-BP1 and inhibit cyclin D1 translation. In contrast, PKCdelta had modest and variable effects on cyclin D1 steady-state levels and failed to restore responsiveness to PKC agonists. Notably, PKCalpha expression blocked anchorage-independent growth in colon cancer cells via a mechanism partially dependent on cyclin D1 deficiency, while PKCdelta had only minor effects. Loss of PKCalpha and effects of its re-expression were independent of the status of the APC/beta-catenin signaling pathway or known genetic alterations, indicating that they are a general characteristic of colon tumors. Thus, PKCalpha is a potent negative regulator of cyclin D1 expression and anchorage-independent cell growth in colon tumor cells, findings that offer important perspectives on the frequent loss of this isozyme during intestinal carcinogenesis.


Subject(s)
Cyclin D1/metabolism , Intestinal Neoplasms/physiopathology , Protein Kinase C/metabolism , Animals , Cell Line, Tumor , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoblotting , Mice , Promoter Regions, Genetic/drug effects , Protein Biosynthesis , Protein Kinase C/pharmacology , Rats , Signal Transduction , Transcription, Genetic
13.
J Cell Physiol ; 218(3): 638-42, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19034928

ABSTRACT

In stimulating maturation of colonic carcinoma cells, the short chain fatty acid butyrate, and 1alpha,25-dihydroxyvitamin D(3), were shown to attenuate transcription of the cyclin D1 gene, giving rise to truncated transcripts of this locus. Moreover, a sequence which is highly conserved in the human, mouse, rat, and dog genome was found in the 4 kb long intron 3 of the human cyclin D1 gene, and is capable of forming a hairpin structure similar to that of microRNA precursors. The expression of this sequence is also decreased by the attenuation. Thus, the transcriptional attenuation at the cyclin D1 locus not only down-regulates the expression of this key gene in mucosal cell maturation and tumorigenesis, but may also abrogate the generation of a molecule that encompasses this conserved sequence in cyclin D1 intron 3.


Subject(s)
Butyrates/pharmacology , Cholecalciferol/pharmacology , Colonic Neoplasms/genetics , Cyclin D1/genetics , Transcription, Genetic/drug effects , Animals , Base Sequence , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Situ Hybridization, Fluorescence , Introns/genetics , Mice , Molecular Sequence Data , Nucleic Acid Conformation
14.
Cancer Res ; 68(19): 7803-10, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829535

ABSTRACT

A defined rodent "new Western diet" (NWD), which recapitulates intake levels of nutrients that are major dietary risk factors for human colon cancer, induced colonic tumors when fed to wild-type C57Bl/6 mice for 1.5 to 2 years from age 6 weeks (two-thirds of their life span). Colonic tumors were prevented by elevating dietary calcium and vitamin D(3) to levels comparable with upper levels consumed by humans, but tumorigenesis was not altered by similarly increasing folate, choline, methionine, or fiber, each of which was also at the lower levels in the NWD that are associated with risk for colon cancer. The NWD significantly altered profiles of gene expression in the flat colonic mucosa that exhibited heterogeneity among the mice, but unsupervised clustering of the data and novel statistical analyses showed reprogramming of colonic epithelial cells in the flat mucosa by the NWD was similar to that initiated by inheritance of a mutant Apc allele. The NWD also caused general down-regulation of genes encoding enzymes involved in lipid metabolism and the tricarboxylic acid cycle in colonic epithelial cells before tumor formation, which was prevented by the supplementation of the NWD with calcium and vitamin D(3) that prevented colon tumor development, demonstrating profound interaction among nutrients. This mouse model of dietary induction of colon cancer recapitulates levels and length of exposure to nutrients linked to relative risk for human sporadic colon cancer, which represents the etiology of >90% of colon cancer in the United States and other Western countries.


Subject(s)
Colonic Neoplasms/etiology , Diet/adverse effects , Disease Models, Animal , Mice , Animals , Cluster Analysis , Colonic Neoplasms/epidemiology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Female , Gene Expression Profiling , Genes, APC , Incidence , Male , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Mucin-1/genetics , Oligonucleotide Array Sequence Analysis , Signal Transduction/genetics
15.
Proc Natl Acad Sci U S A ; 105(39): 15064-9, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18806221

ABSTRACT

We normally live in symbiosis with approximately 10(13) bacteria present in the colon. Among the several mechanisms maintaining the bacteria/host balance, there is limited understanding of the structure, function, and properties of intestinal mucus. We now demonstrate that the mouse colonic mucus consists of two layers extending 150 mum above the epithelial cells. Proteomics revealed that both of these layers have similar protein composition, with the large gel-forming mucin Muc2 as the major structural component. The inner layer is densely packed, firmly attached to the epithelium, and devoid of bacteria. In contrast, the outer layer is movable, has an expanded volume due to proteolytic cleavages of the Muc2 mucin, and is colonized by bacteria. Muc2(-/-) mice have bacteria in direct contact with the epithelial cells and far down in the crypts, explaining the inflammation and cancer development observed in these animals. These findings show that the Muc2 mucin can build a mucus barrier that separates bacteria from the colon epithelia and suggest that defects in this mucus can cause colon inflammation.


Subject(s)
Colon/microbiology , Intestinal Mucosa/microbiology , Mucins/physiology , Mucus/microbiology , Symbiosis , Animals , Colitis/genetics , Colitis/immunology , Colitis/microbiology , Colon/cytology , Colon/immunology , Colon/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Mice, Mutant Strains , Mucin-2 , Mucins/genetics , Mucus/cytology , Mucus/immunology , Mucus/metabolism , Rats , Rats, Sprague-Dawley , Symbiosis/genetics
16.
Cancer Res ; 68(18): 7313-22, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18794118

ABSTRACT

Somatic mutations of the adenomatous polyposis coli (APC) gene are initiating events in the majority of sporadic colon cancers. A common characteristic of such tumors is reduction in the number of goblet cells that produce the mucin MUC2, the principal component of intestinal mucus. Consistent with these observations, we showed that Muc2 deficiency results in the spontaneous development of tumors along the entire gastrointestinal tract, independently of deregulated Wnt signaling. To dissect the complex interaction between Muc2 and Apc in intestinal tumorigenesis and to elucidate the mechanisms of tumor formation in Muc2(-/-) mice, we crossed the Muc2(-/-) mouse with two mouse models, Apc(1638N/+) and Apc(Min/+), each of which carries an inactivated Apc allele. The introduction of mutant Muc2 into Apc(1638N/+) and Apc(Min/+) mice greatly increased transformation induced by the Apc mutation and significantly shifted tumor development toward the colon as a function of Muc2 gene dosage. Furthermore, we showed that in compound double mutant mice, deregulation of Wnt signaling was the dominant mechanism of tumor formation. The increased tumor burden in the distal colon of Muc2/Apc double mutant mice was similar to the phenotype observed in Apc(Min/+) mice that are challenged to mount an inflammatory response, and consistent with this, gene expression profiles of epithelial cells from flat mucosa of Muc2-deficient mice suggested that Muc2 deficiency was associated with low levels of subclinical chronic inflammation. We hypothesize that Muc2(-/-) tumors develop through an inflammation-related pathway that is distinct from and can complement mechanisms of tumorigenesis in Apc(+/-) mice.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genes, APC , Intestinal Neoplasms/genetics , Mucins/genetics , Wnt Proteins/metabolism , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/metabolism , Alleles , Animals , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Enterocolitis/genetics , Enterocolitis/metabolism , Enterocolitis/pathology , Gene Silencing , Immunohistochemistry , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Loss of Heterozygosity , Mice , Mice, Inbred C57BL , Mucin-2 , Mucins/deficiency , Signal Transduction , beta Catenin/metabolism
17.
Lab Invest ; 88(6): 634-42, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18427556

ABSTRACT

Expression of the mucin MUC2, the structural component of the colonic mucus layer, is lowered in ulcerative colitis. Furthermore, interleukin (IL)-10 knockout (IL-10-/-) mice develop colitis and have reduced Muc2 levels. Our aim was to obtain insight into the role of Muc2 and IL-10 in epithelial protection. Muc2-IL-10 double-knockout (Muc2/IL-10(DKO)) mice were characterized and compared to Muc2 knockout (Muc2-/-), IL-10-/- and wild-type (WT) mice. Clinical symptoms, intestinal morphology and differences in epithelial-specific protein levels were analyzed. In addition, levels of the pro-inflammatory cytokines in colonic tissue and serum were determined. IL-10-/- mice were indistinguishable from WT mice throughout this experiment and showed no clinical or histological signs of colitis. Muc2/IL-10(DKO) and Muc2-/- mice showed significant growth retardation and clinical signs of colitis at 4 and 5 weeks, respectively. Muc2/IL-10(DKO) mice had a high mortality rate (50% survival/5 weeks) compared to the other types of mice (100% survival). Microscopic analysis of the colon of Muc2/IL-10(DKO) mice showed mucosal thickening, increased proliferation, superficial erosions and a diminished Muc4 expression. Furthermore, pro-inflammatory cytokines were significantly upregulated, both in tissue (mRNA) and systemically in Muc2/IL-10(DKO) mice. In conclusion, Muc2/IL-10(DKO) mice develop colitis, which is more severe in every aspect compared to Muc2-/- and IL-10-/- mice. These data indicate that (i) in case of Muc2 deficiency, the anti-inflammatory cytokine IL-10 can control epithelial damage, though to a limited extent and (ii) the mucus layer is most likely a key factor determining colitis.


Subject(s)
Epithelium/immunology , Immunologic Factors/metabolism , Inflammation/etiology , Interleukin-10/deficiency , Mucins/deficiency , Animals , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Epithelium/pathology , Heterozygote , Immunohistochemistry , Inflammation/pathology , Interleukin-10/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Mucin-2 , Mucins/genetics
18.
J Cell Physiol ; 210(1): 144-52, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16998802

ABSTRACT

The mucin Muc2 or Mycin2 (Muc2), which is the main structural component of the protective mucus layer, has shown to be upregulated during chemotherapy-induced mucositis. As Muc2 has shown to have protective capacities, upregulation of Muc2 may be a counter reaction of the intestine protecting against mucositis. Therefore, increasing Muc2 protein levels could be a therapeutic target in mucositis prevention or reduction. Our aim was to determine the role of Muc2 in chemotherapy-induced mucositis. Mucositis was induced in Muc2 knockout (Muc2(-/-)) and wild type (Muc2(+/+)) mice by injecting methotrexate (MTX). Animals were weighed and sacrificed on Days 2-6 after MTX treatment and jejunal segments were analyzed. Before MTX treatment, the small intestine of Muc2(+/+) and Muc2(-/-) mice were similar with respect to epithelial morphology and proliferation. Moreover, sucrase-isomaltase and trefoil factor-3 protein expression levels were comparable between Muc2(+/+) and Muc2(-/-) mice. Up to Day 3 after MTX treatment, percentages of weight-loss did not differ. Thereafter, Muc2(+/+) mice showed a trend towards regaining weight, whereas Muc2(-/-) mice continued to lose weight. Surprisingly, MTX-induced intestinal damage of Muc2(-/-) and Muc2(+/+) mice was comparable. Prior to MTX-injection, tumor necrosis factor-alpha and interleukin-10 mRNAs were upregulated in Muc2(-/-) mice, probably due to continuous exposure of the intestine to luminal antigens. Muc2 deficiency does not lead to an increase in chemotherapy-induced mucositis. A possible explanation is the mechanism by which Muc2 deficiency may trigger the immune system to release interleukin-10, an anti-inflammatory cytokine before MTX-treatment.


Subject(s)
Enteritis/pathology , Intestinal Diseases/pathology , Intestines/pathology , Methotrexate/toxicity , Mucins/deficiency , Mucositis/pathology , Animals , Antimetabolites, Antineoplastic/toxicity , Cell Proliferation , Enteritis/chemically induced , Enteritis/metabolism , Enterocytes/metabolism , Goblet Cells/metabolism , Interleukin-10/metabolism , Intestinal Diseases/chemically induced , Intestinal Diseases/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Jejunum/pathology , Mice , Mice, Knockout , Mucin-2 , Mucins/genetics , Mucins/metabolism , Mucositis/chemically induced , Mucositis/metabolism , RNA, Messenger/metabolism , Sucrase-Isomaltase Complex/metabolism , Time Factors , Trefoil Factor-3 , Tumor Necrosis Factor-alpha/metabolism
20.
Gastroenterology ; 131(1): 117-29, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16831596

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.


Subject(s)
Colitis/metabolism , Mucins/metabolism , Animals , Colitis/drug therapy , Colitis/pathology , Dextran Sulfate/therapeutic use , Disease Models, Animal , Gene Expression , Immunohistochemistry , In Situ Hybridization , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mucin-2 , Mucins/deficiency , Mucins/genetics , Plasma Substitutes/therapeutic use , Polymerase Chain Reaction , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...