Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Gastroenterology ; 162(3): 813-827.e8, 2022 03.
Article in English | MEDLINE | ID: mdl-34767785

ABSTRACT

BACKGROUND & AIMS: Because inflammatory bowel disease is increasing worldwide and can lead to colitis-associated carcinoma (CAC), new interventions are needed. We have shown that spermine oxidase (SMOX), which generates spermidine (Spd), regulates colitis. Here we determined whether Spd treatment reduces colitis and carcinogenesis. METHODS: SMOX was quantified in human colitis and associated dysplasia using quantitative reverse-transcription polymerase chain reaction and immunohistochemistry. We used wild-type (WT) and Smox-/- C57BL/6 mice treated with dextran sulfate sodium (DSS) or azoxymethane (AOM)-DSS as models of colitis and CAC, respectively. Mice with epithelial-specific deletion of Apc were used as a model of sporadic colon cancer. Animals were supplemented or not with Spd in the drinking water. Colonic polyamines, inflammation, tumorigenesis, transcriptomes, and microbiomes were assessed. RESULTS: SMOX messenger RNA levels were decreased in human ulcerative colitis tissues and inversely correlated with disease activity, and SMOX protein was reduced in colitis-associated dysplasia. DSS colitis and AOM-DSS-induced dysplasia and tumorigenesis were worsened in Smox-/- vs WT mice and improved in both genotypes with Spd. Tumor development caused by Apc deletion was also reduced by Spd. Smox deletion and AOM-DSS treatment were both strongly associated with increased expression of α-defensins, which was reduced by Spd. A shift in the microbiome, with reduced abundance of Prevotella and increased Proteobacteria and Deferribacteres, occurred in Smox-/- mice and was reversed with Spd. CONCLUSIONS: Loss of SMOX is associated with exacerbated colitis and CAC, increased α-defensin expression, and dysbiosis of the microbiome. Spd supplementation reverses these phenotypes, indicating that it has potential as an adjunctive treatment for colitis and chemopreventive for colon carcinogenesis.


Subject(s)
Carcinogenesis/drug effects , Carcinogenesis/genetics , Colitis/genetics , Colonic Neoplasms/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Spermidine/therapeutic use , Adenomatous Polyposis Coli Protein/genetics , Animals , Azoxymethane , Colitis/chemically induced , Colitis/enzymology , Colitis/prevention & control , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/genetics , Colon/enzymology , Colon/pathology , Colonic Neoplasms/prevention & control , Dextran Sulfate , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation/drug effects , Humans , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Male , Mice , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Precancerous Conditions/enzymology , Protective Factors , RNA, Messenger/metabolism , Severity of Illness Index , Spermidine/metabolism , Spermidine/pharmacology , Weight Loss/drug effects , alpha-Defensins/genetics , alpha-Defensins/metabolism , Polyamine Oxidase
2.
Oncogene ; 40(47): 6540-6546, 2021 11.
Article in English | MEDLINE | ID: mdl-34625710

ABSTRACT

CCL11, also known as eotaxin-1, is described as an eosinophil chemoattractant, which has been implicated in allergic and Th2 inflammatory diseases. We have reported that CCL11 is significantly increased in the serum of inflammatory bowel disease (IBD) patients, colonic eosinophils are increased and correlate with tissue CCL11 levels in ulcerative colitis patients, and CCL11 is increased in dextran sulfate sodium (DSS)-induced murine colitis. Here, we show that CCL11 is involved in the pathogenesis of DSS-induced colitis and in colon tumorigenesis in the azoxymethane (AOM)-DSS model of colitis-associated carcinogenesis (CAC). Ccl11-/- mice exposed to DSS then allowed to recover had significantly less body weight loss and a decrease in histologic injury versus wild-type (WT) mice. In the AOM-DSS model, Ccl11-/- mice exhibited decreased colonic tumor number and burden, histologic injury, and colonic eosinophil infiltration versus WT mice. Ccl11 is expressed by both colonic epithelial and lamina propria immune cells. Studies in bone marrow chimera mice revealed that hematopoietic- and epithelial-cell-derived CCL11 were both important for tumorigenesis in the AOM-DSS model. These findings indicate that CCL11 is important in the regulation of colitis and associated carcinogenesis and thus anti-CCL11 antibodies may be useful for treatment and cancer chemoprevention in IBD.


Subject(s)
Carcinogenesis/pathology , Chemokine CCL11/physiology , Colitis-Associated Neoplasms/pathology , Colitis/complications , Epithelial Cells/pathology , Animals , Azoxymethane/toxicity , Carcinogenesis/metabolism , Carcinogens/toxicity , Colitis/chemically induced , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/metabolism , Epithelial Cells/metabolism , Mice , Mice, Knockout
3.
Mucosal Immunol ; 14(5): 1088-1099, 2021 09.
Article in English | MEDLINE | ID: mdl-34183776

ABSTRACT

CD4+ T cell activation and differentiation are important events that set the stage for proper immune responses. Many factors are involved in the activation and differentiation of T cells, and these events are tightly controlled to prevent unwanted and/or exacerbated immune responses that may harm the host. It has been well-documented that granzyme B, a potent serine protease involved in cell-mediated cytotoxicity, is readily expressed by certain CD4+ T cells, such as regulatory T cells and CD4+CD8αα+ intestinal intraepithelial lymphocytes, both of which display cytotoxicity associated with granzyme B. However, because not all CD4+ T cells expressing granzyme B are cytotoxic, additional roles for this protease in CD4+ T cell biology remain unknown. Here, using a combination of in vivo and in vitro approaches, we report that granzyme B-deficient CD4+ T cells display increased IL-17 production. In the adoptive transfer model of intestinal inflammation, granzyme B-deficient CD4+ T cells triggered a more rapid disease onset than their WT counterparts, and presented a differential transcription profile. Similar results were also observed in granzyme B-deficient mice infected with Citrobacter rodentium. Our results suggest that granzyme B modulates CD4+ T cell differentiation, providing a new perspective into the biology of this enzyme.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Disease Susceptibility , Granzymes/genetics , Interleukin-17/biosynthesis , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Animals , Biomarkers , Cell Differentiation/immunology , Cell Transplantation , Cytokines/biosynthesis , Female , Gene Expression Profiling , Gene Expression Regulation , Granzymes/metabolism , Immune Reconstitution , Immunophenotyping , Lymphocyte Activation , Male , Mice , Mice, Knockout , Mice, Transgenic , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
4.
Gastroenterology ; 160(4): 1256-1268.e9, 2021 03.
Article in English | MEDLINE | ID: mdl-33189701

ABSTRACT

BACKGROUND & AIMS: Inflammation in the gastrointestinal tract may lead to the development of cancer. Dicarbonyl electrophiles, such as isolevuglandins (isoLGs), are generated from lipid peroxidation during the inflammatory response and form covalent adducts with amine-containing macromolecules. Thus, we sought to determine the role of dicarbonyl electrophiles in inflammation-associated carcinogenesis. METHODS: The formation of isoLG adducts was analyzed in the gastric tissues of patients infected with Helicobacter pylori from gastritis to precancerous intestinal metaplasia, in human gastric organoids, and in patients with colitis and colitis-associated carcinoma (CAC). The effect on cancer development of a potent scavenger of dicarbonyl electrophiles, 5-ethyl-2-hydroxybenzylamine (EtHOBA), was determined in transgenic FVB/N insulin-gastrin (INS-GAS) mice and Mongolian gerbils as models of H pylori-induced carcinogenesis and in C57BL/6 mice treated with azoxymethane-dextran sulfate sodium as a model of CAC. The effect of EtHOBA on mutations in gastric epithelial cells of H pylori-infected INS-GAS mice was assessed by whole-exome sequencing. RESULTS: We show increased isoLG adducts in gastric epithelial cell nuclei in patients with gastritis and intestinal metaplasia and in human gastric organoids infected with H pylori. EtHOBA inhibited gastric carcinoma in infected INS-GAS mice and gerbils and attenuated isoLG adducts, DNA damage, and somatic mutation frequency. Additionally, isoLG adducts were elevated in tissues from patients with colitis, colitis-associated dysplasia, and CAC as well as in dysplastic tumors of C57BL/6 mice treated with azoxymethane-dextran sulfate sodium. In this model, EtHOBA significantly reduced adduct formation, tumorigenesis, and dysplasia severity. CONCLUSIONS: Dicarbonyl electrophiles represent a link between inflammation and somatic genomic alterations and are thus key targets for cancer chemoprevention.


Subject(s)
Cell Transformation, Neoplastic/immunology , Colitis-Associated Neoplasms/immunology , Lipids/immunology , Precancerous Conditions/immunology , Stomach Neoplasms/immunology , Animals , Benzylamines/pharmacology , Benzylamines/therapeutic use , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/drug effects , Colitis-Associated Neoplasms/microbiology , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/prevention & control , Disease Models, Animal , Epithelial Cells , Gastric Mucosa/cytology , Gastric Mucosa/drug effects , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Gastritis/immunology , Gastritis/microbiology , Gastritis/pathology , Gerbillinae , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/immunology , Helicobacter pylori/isolation & purification , Humans , Lipids/antagonists & inhibitors , Metaplasia/immunology , Metaplasia/microbiology , Metaplasia/pathology , Mice , Mice, Transgenic , Organoids , Precancerous Conditions/drug therapy , Precancerous Conditions/microbiology , Precancerous Conditions/pathology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Stomach Neoplasms/prevention & control
5.
Article in English | MEDLINE | ID: mdl-30972302

ABSTRACT

There is great interest in safe and effective alternative therapies that could benefit patients with inflammatory bowel diseases (IBD). L-arginine (Arg) is a semi-essential amino acid with a variety of physiological effects. In this context, our aim was to investigate the role of dietary Arg in experimental colitis. We used two models of colitis in C57BL/6 mice, the dextran sulfate sodium (DSS) model of injury and repair, and Citrobacter rodentium infection. Animals were given diets containing (1) no Arg (Arg0), 6.4 g/kg (ArgNL), or 24.6 g/kg Arg (ArgHIGH); or (2) the amino acids downstream of Arg: 28 g/kg L-ornithine (OrnHIGH) or 72 g/kg L-proline (ProHIGH). Mice with DSS colitis receiving the ArgHIGH diet had increased levels of Arg, Orn, and Pro in the colon and improved body weight loss, colon length shortening, and histological injury compared to ArgNL and Arg0 diets. Histology was improved in the ArgNL vs. Arg0 group. OrnHIGH or ProHIGH diets did not provide protection. Reduction in colitis with ArgHIGH diet also occurred in C. rodentium-infected mice. Diversity of the intestinal microbiota was significantly enhanced in mice on the ArgHIGH diet compared to the ArgNL or Arg0 diets, with increased abundance of Bacteroidetes and decreased Verrucomicrobia. In conclusion, dietary supplementation of Arg is protective in colitis models. This may occur by restoring overall microbial diversity and Bacteroidetes prevalence. Our data provide a rationale for Arg as an adjunctive therapy in IBD.


Subject(s)
Arginine/administration & dosage , Colitis/pathology , Colon/microbiology , Diet/methods , Enterobacteriaceae Infections/pathology , Gastrointestinal Microbiome , Animals , Citrobacter rodentium/growth & development , Colitis/chemically induced , Colon/pathology , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Disease Models, Animal , Histocytochemistry , Mice, Inbred C57BL , Treatment Outcome
6.
Oncogene ; 38(7): 1067-1079, 2019 02.
Article in English | MEDLINE | ID: mdl-30202097

ABSTRACT

Solute carrier family 7 member 2 (SLC7A2, also known as CAT2) is an inducible transporter of the semi-essential amino acid L-arginine (L-Arg), which has been implicated in wound repair. We have reported that both SLC7A2 expression and L-Arg availability are decreased in colonic tissues from inflammatory bowel disease patients and that mice lacking Slc7a2 exhibit a more severe disease course when exposed to dextran sulfate sodium (DSS) compared to wild-type (WT) mice. Here, we present evidence that SLC7A2 plays a role in modulating colon tumorigenesis in the azoxymethane (AOM)-DSS model of colitis-associated carcinogenesis (CAC). SLC7A2 was localized predominantly to colonic epithelial cells in WT mice. Utilizing the AOM-DSS model, Slc7a2-/- mice had significantly increased tumor number, burden, and risk of high-grade dysplasia vs. WT mice. Tumors from Slc7a2-/- mice exhibited significantly increased levels of the proinflammatory cytokines/chemokines IL-1ß, CXCL1, CXCL5, IL-3, CXCL2, CCL3, and CCL4, but decreased levels of IL-4, CXCL9, and CXCL10 compared to tumors from WT mice. This was accompanied by a shift toward pro-tumorigenic M2 macrophage activation in Slc7a2-deficient mice, as marked by increased colonic CD11b+F4/80+ARG1+ cells with no alteration in CD11b+F4/80+NOS2+ cells by flow cytometry and immunofluorescence microscopy. The shift toward M2 macrophage activation was confirmed in bone marrow-derived macrophages from Slc7a2-/- mice. In bone marrow chimeras between Slc7a2-/- and WT mice, the recipient genotype drove the CAC phenotype, suggesting the importance of epithelial SLC7A2 in abrogating neoplastic risk. These data reveal that SLC7A2 has a significant role in the protection from CAC in the setting of chronic colitis, and suggest that the decreased SLC7A2 in inflammatory bowel disease (IBD) may contribute to CAC risk. Strategies to enhance L-Arg availability by supplementing L-Arg and/or increasing L-Arg uptake could represent a therapeutic approach in IBD to reduce the substantial long-term risk of colorectal carcinoma.


Subject(s)
Amino Acid Transport Systems, Basic/metabolism , Cell Transformation, Neoplastic/metabolism , Colonic Neoplasms/metabolism , Neoplasm Proteins/metabolism , Amino Acid Transport Systems, Basic/genetics , Animals , Azoxymethane/toxicity , Cell Line, Tumor , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics
7.
Cancer Res ; 78(15): 4303-4315, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29853605

ABSTRACT

Ornithine decarboxylase (ODC) is the rate-limiting enzyme for polyamine biosynthesis and restricts M1 macrophage activation in gastrointestinal (GI) infections. However, the role of macrophage ODC in colonic epithelial-driven inflammation is unknown. Here, we investigate cell-specific effects of ODC in colitis and colitis-associated carcinogenesis (CAC). Human colonic macrophages expressed increased ODC levels in active ulcerative colitis and Crohn's disease, colitis-associated dysplasia, and CAC. Mice lacking Odc in myeloid cells (OdcΔmye mice) that were treated with dextran sulfate sodium (DSS) exhibited improved survival, body weight, and colon length and reduced histologic injury versus control mice. In contrast, GI epithelial-specific Odc knockout had no effect on clinical parameters. Despite reduced histologic damage, colitis tissues of OdcΔmye mice had increased levels of multiple proinflammatory cytokines and chemokines and enhanced expression of M1, but not M2 markers. In the azoxymethane-DSS model of CAC, OdcΔmye mice had reduced tumor number, burden, and high-grade dysplasia. Tumors from OdcΔmye mice had increased M1, but not M2 macrophages. Increased levels of histone 3, lysine 9 acetylation, a marker of open chromatin, were manifest in tumor macrophages of OdcΔmye mice, consistent with our findings that macrophage ODC affects histone modifications that upregulate M1 gene transcription during GI infections. These findings support the concept that macrophage ODC augments epithelial injury-associated colitis and CAC by impairing the M1 responses that stimulate epithelial repair, antimicrobial defense, and antitumoral immunity. They also suggest that macrophage ODC is an important target for colon cancer chemoprevention.Significance: Ornithine decarboxylase contributes to the pathogenesis of colitis and associated carcinogenesis by impairing M1 macrophage responses needed for antitumoral immunity; targeting ODC in macrophages may represent a new strategy for chemoprevention. Cancer Res; 78(15); 4303-15. ©2018 AACR.


Subject(s)
Carcinogenesis/immunology , Colitis, Ulcerative/immunology , Colon/immunology , Colonic Neoplasms/immunology , Macrophages/immunology , Ornithine Decarboxylase/immunology , Animals , Azoxymethane/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/pathology , Colitis, Ulcerative/pathology , Colon/pathology , Colonic Neoplasms/pathology , Cytokines/immunology , Dextran Sulfate/pharmacology , Inflammation/immunology , Inflammation/pathology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophage Activation/physiology , Macrophages/drug effects , Male , Mice , Transcription, Genetic/drug effects , Transcription, Genetic/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
8.
Mucosal Immunol ; 11(5): 1363-1374, 2018 09.
Article in English | MEDLINE | ID: mdl-29907869

ABSTRACT

Blood vessel epicardial substance (BVES), or POPDC1, is a tight junction-associated transmembrane protein that modulates epithelial-to-mesenchymal transition (EMT) via junctional signaling pathways. There have been no in vivo studies investigating the role of BVES in colitis. We hypothesized that BVES is critical for maintaining colonic epithelial integrity. At baseline, Bves-/- mouse colons demonstrate increased crypt height, elevated proliferation, decreased apoptosis, altered intestinal lineage allocation, and dysregulation of tight junctions with functional deficits in permeability and altered intestinal immunity. Bves-/- mice inoculated with Citrobacter rodentium had greater colonic injury, increased colonic and mesenteric lymph node bacterial colonization, and altered immune responses after infection. We propose that increased bacterial colonization and translocation result in amplified immune responses and worsened injury. Similarly, dextran sodium sulfate (DSS) treatment resulted in greater histologic injury in Bves-/- mice. Two different human cell lines (Caco2 and HEK293Ts) co-cultured with enteropathogenic E. coli showed increased attaching/effacing lesions in the absence of BVES. Finally, BVES mRNA levels were reduced in human ulcerative colitis (UC) biopsy specimens. Collectively, these studies suggest that BVES plays a protective role both in ulcerative and infectious colitis and identify BVES as a critical protector of colonic mucosal integrity.


Subject(s)
Colitis, Ulcerative/metabolism , Colon/metabolism , Epithelial Cells/metabolism , Intestinal Absorption/physiology , Membrane Proteins/metabolism , Adult , Animals , Caco-2 Cells , Cell Adhesion Molecules , Cell Line , Cell Line, Tumor , Citrobacter rodentium/pathogenicity , Coculture Techniques , Colon/drug effects , Dextran Sulfate/pharmacology , Epithelial Cells/drug effects , Escherichia coli/metabolism , Female , HEK293 Cells , Humans , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Muscle Proteins , Permeability/drug effects , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tight Junctions/drug effects , Tight Junctions/metabolism
9.
Front Immunol ; 9: 1242, 2018.
Article in English | MEDLINE | ID: mdl-29922289

ABSTRACT

Polyamines have been implicated in numerous biological processes, including inflammation and carcinogenesis. Homeostatic regulation leads to interconversion of the polyamines putrescine and the downstream metabolites spermidine and spermine. The enzyme spermine oxidase (SMOX), which back-converts spermine to spermidine, contributes to regulation of polyamine levels, but can also have other effects. We have implicated SMOX in gastric inflammation and carcinogenesis due to infection by the pathogen Helicobacter pylori. In addition, we reported that SMOX can be upregulated in humans with inflammatory bowel disease. Herein, we utilized Smox-deficient mice to examine the role of SMOX in two murine colitis models, Citrobacter rodentium infection and dextran sulfate sodium (DSS)-induced epithelial injury. In C. rodentium-infected wild-type (WT) mice, there were marked increases in colon weight/length and histologic injury, with mucosal hyperplasia and inflammatory cell infiltration; these changes were ameliorated in Smox-/- mice. In contrast, with DSS, Smox-/- mice exhibited substantial mortality, and increased body weight loss, colon weight/length, and histologic damage. In C. rodentium-infected WT mice, there were increased colonic levels of the chemokines CCL2, CCL3, CCL4, CXCL1, CXCL2, and CXCL10, and the cytokines IL-6, TNF-α, CSF3, IFN-γ, and IL-17; each were downregulated in Smox-/- mice. In DSS colitis, increased levels of IL-6, CSF3, and IL-17 were further increased in Smox-/- mice. In both models, putrescine and spermidine were increased in WT mice; in Smox-/- mice, the main effect was decreased spermidine and spermidine/spermine ratio. With C. rodentium, polyamine levels correlated with histologic injury, while with DSS, spermidine was inversely correlated with injury. Our studies indicate that SMOX has immunomodulatory effects in experimental colitis via polyamine flux. Thus, SMOX contributes to the immunopathogenesis of C. rodentium infection, but is protective in DSS colitis, indicating the divergent effects of spermidine.


Subject(s)
Colitis/etiology , Colitis/metabolism , Immunomodulation , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Animals , Citrobacter rodentium/physiology , Colitis/pathology , Cytokines/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Gene Deletion , Immunity, Mucosal/genetics , Immunomodulation/drug effects , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Oxidoreductases Acting on CH-NH Group Donors/genetics , Spermidine/metabolism , Spermidine/pharmacology , Spermine/metabolism , Spermine/pharmacology , Polyamine Oxidase
10.
Proc Natl Acad Sci U S A ; 114(5): E751-E760, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096401

ABSTRACT

Macrophage activation is a critical step in host responses during bacterial infections. Ornithine decarboxylase (ODC), the rate-limiting enzyme in polyamine metabolism, has been well studied in epithelial cells and is known to have essential roles in many different cellular functions. However, its role in regulating macrophage function during bacterial infections is not well characterized. We demonstrate that macrophage-derived ODC is a critical regulator of M1 macrophage activation during both Helicobacter pylori and Citrobacter rodentium infection. Myeloid-specific Odc deletion significantly increased gastric and colonic inflammation, respectively, and enhanced M1 activation. Add-back of putrescine, the product of ODC, reversed the increased macrophage activation, indicating that ODC and putrescine are regulators of macrophage function. Odc-deficient macrophages had increased histone 3, lysine 4 (H3K4) monomethylation, and H3K9 acetylation, accompanied by decreased H3K9 di/trimethylation both in vivo and ex vivo in primary macrophages. These alterations in chromatin structure directly resulted in up-regulated gene transcription, especially M1 gene expression. Thus, ODC in macrophages tempers antimicrobial, M1 macrophage responses during bacterial infections through histone modifications and altered euchromatin formation, leading to the persistence and pathogenesis of these organisms.


Subject(s)
Enterobacteriaceae Infections/immunology , Helicobacter Infections/immunology , Histones/metabolism , Macrophages/immunology , Ornithine Decarboxylase/immunology , Animals , Cell Line , Citrobacter rodentium , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/pathology , Cytokines/immunology , Enterobacteriaceae Infections/pathology , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Gastritis/immunology , Gastritis/pathology , Helicobacter Infections/pathology , Helicobacter pylori , Humans , Macrophage Activation , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Ornithine Decarboxylase/genetics , Putrescine/metabolism
11.
PLoS Pathog ; 12(10): e1005984, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27783672

ABSTRACT

Solute carrier family 7 member 2 (SLC7A2) is an inducible transporter of the semi-essential amino acid L-arginine (L-Arg), which has been implicated in immune responses to pathogens. We assessed the role of SLC7A2 in murine infection with Citrobacter rodentium, an attaching and effacing enteric pathogen that causes colitis. Induction of SLC7A2 was upregulated in colitis tissues, and localized predominantly to colonic epithelial cells. Compared to wild-type mice, Slc7a2-/-mice infected with C. rodentium had improved survival and decreased weight loss, colon weight, and histologic injury; this was associated with decreased colonic macrophages, dendritic cells, granulocytes, and Th1 and Th17 cells. In infected Slc7a2-/-mice, there were decreased levels of the proinflammatory cytokines G-CSF, TNF-α, IL-1α, IL-1ß, and the chemokines CXCL1, CCL2, CCL3, CCL4, CXCL2, and CCL5. In bone marrow chimeras, the recipient genotype drove the colitis phenotype, indicative of the importance of epithelial, rather than myeloid SLC7A2. Mice lacking Slc7a2 exhibited reduced adherence of C. rodentium to the colonic epithelium and decreased expression of Talin-1, a focal adhesion protein involved in the attachment of the bacterium. The importance of SLC7A2 and Talin-1 in the intimate attachment of C. rodentium and induction of inflammatory response was confirmed in vitro, using conditionally-immortalized young adult mouse colon (YAMC) cells with shRNA knockdown of Slc7a2 or Tln1. Inhibition of L-Arg uptake with the competitive inhibitor, L-lysine (L-Lys), also prevented attachment of C. rodentium and chemokine expression. L-Lys and siRNA knockdown confirmed the role of L-Arg and SLC7A2 in human Caco-2 cells co-cultured with enteropathogenic Escherichia coli. Overexpression of SLC7A2 in human embryonic kidney cells increased bacterial adherence and chemokine expression. Taken together, our data indicate that C. rodentium enhances its own pathogenicity by inducing the expression of SLC7A2 to favor its attachment to the epithelium and thus create its ecological niche.


Subject(s)
Cationic Amino Acid Transporter 2/metabolism , Enterobacteriaceae Infections/metabolism , Host-Parasite Interactions/physiology , Animals , Blotting, Western , Cationic Amino Acid Transporter 2/immunology , Cell Line , Citrobacter rodentium , Disease Models, Animal , Enterobacteriaceae Infections/immunology , Humans , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Knockout , Transfection
12.
J Clin Invest ; 126(9): 3296-312, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27482886

ABSTRACT

EGFR signaling regulates macrophage function, but its role in bacterial infection has not been investigated. Here, we assessed the role of macrophage EGFR signaling during infection with Helicobacter pylori, a bacterial pathogen that causes persistent inflammation and gastric cancer. EGFR was phosphorylated in murine and human macrophages during H. pylori infection. In human gastric tissues, elevated levels of phosphorylated EGFR were observed throughout the histologic cascade from gastritis to carcinoma. Deleting Egfr in myeloid cells attenuated gastritis and increased H. pylori burden in infected mice. EGFR deficiency also led to a global defect in macrophage activation that was associated with decreased cytokine, chemokine, and NO production. We observed similar alterations in macrophage activation and disease phenotype in the Citrobacter rodentium model of murine infectious colitis. Mechanistically, EGFR signaling activated NF-κB and MAPK1/3 pathways to induce cytokine production and macrophage activation. Although deletion of Egfr had no effect on DC function, EGFR-deficient macrophages displayed impaired Th1 and Th17 adaptive immune responses to H. pylori, which contributed to decreased chronic inflammation in infected mice. Together, these results indicate that EGFR signaling is central to macrophage function in response to enteric bacterial pathogens and is a potential therapeutic target for infection-induced inflammation and associated carcinogenesis.


Subject(s)
Bacterial Infections/metabolism , ErbB Receptors/metabolism , Macrophage Activation , Macrophages/metabolism , Animals , Chemokines/metabolism , Citrobacter rodentium , Cytokines/metabolism , Disease Progression , Female , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori , Humans , Inflammation , Male , Mice , Mice, Transgenic , NF-kappa B/metabolism , Phosphorylation , Signal Transduction
13.
Am J Physiol Gastrointest Liver Physiol ; 308(6): G562-71, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25573176

ABSTRACT

Myeloid translocation genes (MTGs) are transcriptional corepressors implicated in development, malignancy, differentiation, and stem cell function. While MTG16 loss renders mice sensitive to chemical colitis, the role of MTG16 in the small intestine is unknown. Histological examination revealed that Mtg16(-/-) mice have increased enterocyte proliferation and goblet cell deficiency. After exposure to radiation, Mtg16(-/-) mice exhibited increased crypt viability and decreased apoptosis compared with wild-type (WT) mice. Flow cytometric and immunofluorescence analysis of intestinal epithelial cells for phospho-histone H2A.X also indicated decreased DNA damage and apoptosis in Mtg16(-/-) intestines. To determine if Mtg16 deletion affected epithelial cells in a cell-autonomous fashion, intestinal crypts were isolated from Mtg16(-/-) mice. Mtg16(-/-) and WT intestinal crypts showed similar enterosphere forming efficiencies when cultured in the presence of EGF, Noggin, and R-spondin. However, when Mtg16(-/-) crypts were cultured in the presence of Wnt3a, they demonstrated higher enterosphere forming efficiencies and delayed progression to mature enteroids. Mtg16(-/-) intestinal crypts isolated from irradiated mice exhibited increased survival compared with WT intestinal crypts. Interestingly, Mtg16 expression was reduced in a stem cell-enriched population at the time of crypt regeneration. This is consistent with MTG16 negatively regulating regeneration in vivo. Taken together, our data demonstrate that MTG16 loss promotes radioresistance and impacts intestinal stem cell function, possibly due to shifting cellular response away from DNA damage-induced apoptosis and towards DNA repair after injury.


Subject(s)
Cell Proliferation , Gamma Rays , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Nuclear Proteins/metabolism , Radiation Injuries, Experimental/metabolism , Regeneration , Transcription Factors/metabolism , Animals , Apoptosis , Cell Proliferation/drug effects , Cell Survival , DNA Damage , Female , Gene Expression Regulation , Goblet Cells/metabolism , Goblet Cells/pathology , Histones/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestine, Small/drug effects , Intestine, Small/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phenotype , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/pathology , Radiation Tolerance , Regeneration/drug effects , Repressor Proteins , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology , Tissue Culture Techniques , Transcription Factors/deficiency , Transcription Factors/genetics , Wnt3A Protein/pharmacology
14.
Immunity ; 41(3): 451-464, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25220211

ABSTRACT

Innate immune responses are critical for mucosal immunity. Here we describe an innate lymphocyte population, iCD8α cells, characterized by expression of CD8α homodimers. iCD8α cells exhibit innate functional characteristics such as the capacity to engulf and kill bacteria. Development of iCD8α cells depends on expression of interleukin-2 receptor γ chain (IL-2Rγc), IL-15, and the major histocompatibility complex (MHC) class Ib protein H2-T3, also known as the thymus leukemia antigen or TL. While lineage tracking experiments indicated that iCD8α cells have a lymphoid origin, their development was independent of the transcriptional suppressor Id2, suggesting that these cells do not belong to the family of innate lymphoid cells. Finally, we identified cells with a similar phenotype in humans, which were profoundly depleted in newborns with necrotizing enterocolitis. These findings suggest a critical role of iCD8α cells in immune responses associated with the intestinal epithelium.


Subject(s)
Antigen Presentation/immunology , CD8 Antigens/biosynthesis , Immunity, Mucosal/immunology , Intestinal Mucosa/cytology , Lymphocytes/immunology , Animals , Citrobacter rodentium/immunology , Cytochalasin D/pharmacology , Enterocolitis, Necrotizing , Helicobacter pylori/immunology , Histocompatibility Antigens Class I/biosynthesis , Humans , Inhibitor of Differentiation Protein 2/genetics , Interleukin Receptor Common gamma Subunit/biosynthesis , Interleukin-15/biosynthesis , Interleukin-2/biosynthesis , Interleukin-7/biosynthesis , Intestinal Mucosa/immunology , Lymphocyte Activation/immunology , Lymphocytes/classification , Lymphocytes/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/drug effects , Phagocytosis/immunology
15.
PLoS One ; 8(12): e82300, 2013.
Article in English | MEDLINE | ID: mdl-24367513

ABSTRACT

Accurate and high-throughput technologies are needed for identification of new therapeutic targets and for optimizing therapy in inflammatory bowel disease. Our aim was to assess multi-analyte protein-based assays of cytokines/chemokines using Luminex technology. We have reported that Luminex-based profiling was useful in assessing response to L-arginine therapy in the mouse model of dextran sulfate sodium colitis. Therefore, we studied prospectively collected samples from ulcerative colitis (UC) patients and control subjects. Serum, colon biopsies, and clinical information were obtained from subjects undergoing colonoscopy for evaluation of UC or for non-UC indications. In total, 38 normal controls and 137 UC cases completed the study. Histologic disease severity and the Mayo Disease Activity Index (DAI) were assessed. Serum and colonic tissue cytokine/chemokine profiles were measured by Luminex-based multiplex testing of 42 analytes. Only eotaxin-1 and G-CSF were increased in serum of patients with histologically active UC vs. controls. While 13 cytokines/chemokines were increased in active UC vs. controls in tissues, only eotaxin-1 was increased in all levels of active disease in both serum and tissue. In tissues, eotaxin-1 correlated with the DAI and with eosinophil counts. Increased eotaxin-1 levels were confirmed by real-time PCR. Tissue eotaxin-1 levels were also increased in experimental murine colitis induced by dextran sulfate sodium, oxazolone, or Citrobacter rodentium, but not in murine Helicobacter pylori infection. Our data implicate eotaxin-1 as an etiologic factor and therapeutic target in UC, and indicate that Luminex-based assays may be useful to assess IBD pathogenesis and to select patients for anti-cytokine/chemokine therapies.


Subject(s)
Chemokine CCL11/metabolism , Colitis, Ulcerative/metabolism , Adult , Animals , Colitis, Ulcerative/blood , Colitis, Ulcerative/chemically induced , Female , Gastritis/blood , Gastritis/metabolism , Gastritis/microbiology , Helicobacter pylori/pathogenicity , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Oxazolone/toxicity , Prospective Studies
16.
PLoS One ; 8(7): e67845, 2013.
Article in English | MEDLINE | ID: mdl-23861820

ABSTRACT

Selenium (Se) is an essential micronutrient that exerts its functions via selenoproteins. Little is known about the role of Se in inflammatory bowel disease (IBD). Epidemiological studies have inversely correlated nutritional Se status with IBD severity and colon cancer risk. Moreover, molecular studies have revealed that Se deficiency activates WNT signaling, a pathway essential to intestinal stem cell programs and pivotal to injury recovery processes in IBD that is also activated in inflammatory neoplastic transformation. In order to better understand the role of Se in epithelial injury and tumorigenesis resulting from inflammatory stimuli, we examined colonic phenotypes in Se-deficient or -sufficient mice in response to dextran sodium sulfate (DSS)-induced colitis, and azoxymethane (AOM) followed by cyclical administration of DSS, respectively. In response to DSS alone, Se-deficient mice demonstrated increased morbidity, weight loss, stool scores, and colonic injury with a concomitant increase in DNA damage and increases in inflammation-related cytokines. As there was an increase in DNA damage as well as expression of several EGF and TGF-ß pathway genes in response to inflammatory injury, we sought to determine if tumorigenesis was altered in the setting of inflammatory carcinogenesis. Se-deficient mice subjected to AOM/DSS treatment to model colitis-associated cancer (CAC) had increased tumor number, though not size, as well as increased incidence of high grade dysplasia. This increase in tumor initiation was likely due to a general increase in colonic DNA damage, as increased 8-OHdG staining was seen in Se-deficient tumors and adjacent, non-tumor mucosa. Taken together, our results indicate that Se deficiency worsens experimental colitis and promotes tumor development and progression in inflammatory carcinogenesis.


Subject(s)
Carcinogenesis/metabolism , Colitis/metabolism , Colonic Neoplasms/metabolism , Selenium/deficiency , 8-Hydroxy-2'-Deoxyguanosine , Animals , Azoxymethane , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Carcinogenesis/immunology , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , DNA Damage , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/chemistry , Dextran Sulfate , Diet , Epidermal Growth Factor/genetics , Epidermal Growth Factor/immunology , Gene Expression Regulation , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Weight Loss
17.
Am J Physiol Gastrointest Liver Physiol ; 305(3): G225-40, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23703655

ABSTRACT

L-Arginine (L-Arg) is a semiessential amino acid that has altered availability in human ulcerative colitis (UC), a form of inflammatory bowel disease, and is beneficial in murine colitis induced by dextran sulfate sodium (DSS), a model with similarity to UC. We assessed the role of cationic amino acid transporter 2 (CAT2), the inducible transporter of L-Arg, in DSS colitis. Expression of CAT2 was upregulated in tissues from colitic mice and localized predominantly to colonic macrophages. CAT2-deficient (CAT2-/-) mice exposed to DSS exhibited worsening of survival, body weight loss, colon weight, and histological injury. These effects were associated with increased serum L-Arg and decreased tissue L-Arg uptake and inducible nitric oxide synthase protein expression. Clinical benefits of L-Arg supplementation in wild-type mice were lost in CAT2-/- mice. There was increased infiltration of macrophages, dendritic cells, granulocytes, and T cells in colitic CAT2-/- compared with wild-type mice. Cytokine profiling revealed increases in proinflammatory granulocyte colony-stimulating factor, macrophage inflammatory protein-1α, IL-15, and regulated and normal T cell-expressed and -secreted and a shift from an IFN-γ- to an IL-17-predominant T cell response, as well as an increase in IL-13, in tissues from colitic CAT2-/- mice. However, there were no increases in other T helper cell type 2 cytokines, nor was there a global increase in macrophage-derived proinflammatory cytokines. The increase in IL-17 derived from both CD4 and γδ T cells and was associated with colonic IL-6 expression. Thus CAT2 plays an important role in controlling inflammation and IL-17 activation in an injury model of colitis, and impaired L-Arg availability may contribute to UC pathogenesis.


Subject(s)
Cationic Amino Acid Transporter 2/deficiency , Colitis/chemically induced , Colitis/immunology , Dextran Sulfate , Interleukin-17/metabolism , T-Lymphocytes/immunology , Animals , Arginine/metabolism , Cationic Amino Acid Transporter 2/genetics , Cationic Amino Acid Transporter 2/physiology , Colitis/physiopathology , Interleukin-17/genetics , Interleukin-23/genetics , Interleukin-6/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/analysis , Up-Regulation
18.
Gut ; 62(10): 1446-55, 2013 Oct.
Article in English | MEDLINE | ID: mdl-22833394

ABSTRACT

OBJECTIVE: The myeloid translocation genes (MTGs) are transcriptional corepressors with both Mtg8(-/-) and Mtgr1(-/-) mice showing developmental and/or differentiation defects in the intestine. We sought to determine the role of MTG16 in intestinal integrity. METHODS: Baseline and stress induced colonic phenotypes were examined in Mtg16(-/-) mice. To unmask phenotypes, we treated Mtg16(-/-) mice with dextran sodium sulphate (DSS) or infected them with Citrobacter rodentium and the colons were examined for ulceration and for changes in proliferation, apoptosis and inflammation. RESULTS: Mtg16(-/-) mice have altered immune subsets, suggesting priming towards Th1 responses. Mtg16(-/-) mice developed increased weight loss, diarrhoea, mortality and histological colitis and there were increased innate (Gr1(+), F4/80(+), CD11c(+) and MHCII(+); CD11c(+)) and Th1 adaptive (CD4) immune cells in Mtg16(-/-) colons after DSS treatment. Additionally, there was increased apoptosis and a compensatory increased proliferation in Mtg16(-/-) colons. Compared with wild-type mice, Mtg16(-/-) mice exhibited increased colonic CD4;IFN-γ cells in vehicle-treated and DSS-treated mice. Adoptive transfer of wild-type marrow into Mtg16(-/-) recipients did not rescue the Mtg16(-/-) injury phenotype. Isolated colonic epithelial cells from DSS-treated Mtg16(-/-) mice exhibited increased KC (Cxcl1) mRNA expression when compared with wild-type mice. Mtg16(-/-) mice infected with C rodentium had more severe colitis and greater bacterial colonisation. Last, MTG16 mRNA levels were reduced in human ulcerative colitis versus normal colon tissues. CONCLUSIONS: These observations indicate that MTG16 is critical for colonocyte survival and regeneration in response to intestinal injury and provide evidence that this transcriptional corepressor regulates inflammatory recruitment in response to injury.


Subject(s)
Colitis/pathology , Nuclear Proteins/physiology , Transcription Factors/physiology , Adaptive Immunity , Adoptive Transfer , Animals , Bone Transplantation , Cell Proliferation , Colitis/chemically induced , Colitis/immunology , Colitis/physiopathology , Colitis, Ulcerative/metabolism , Colon/immunology , Dextran Sulfate , Enterocytes/pathology , Female , Humans , Immunity, Innate , Immunophenotyping , Intestinal Absorption/physiology , Intestinal Mucosa/pathology , Intestinal Mucosa/physiopathology , Male , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Permeability , Repressor Proteins , Th1 Cells/immunology , Transcription Factors/deficiency , Transcription Factors/genetics
19.
PLoS One ; 7(3): e33546, 2012.
Article in English | MEDLINE | ID: mdl-22428068

ABSTRACT

Inflammatory bowel disease (IBD), consisting of Crohn's disease and ulcerative colitis (UC), results in substantial morbidity and is difficult to treat. New strategies for adjunct therapies are needed. One candidate is the semi-essential amino acid, L-arginine (L-Arg), a complementary medicine purported to be an enhancer of immunity and vitality in the lay media. Using dextran sulfate sodium (DSS) as a murine colonic injury and repair model with similarities to human UC, we assessed the effect of L-Arg, as DSS induced increases in colonic expression of the y(+) cationic amino acid transporter 2 (CAT2) and L-Arg uptake. L-Arg supplementation improved the clinical parameters of survival, body weight loss, and colon weight, and reduced colonic permeability and the number of myeloperoxidase-positive neutrophils in DSS colitis. Luminex-based multi-analyte profiling demonstrated that there was a marked reduction in proinflammatory cytokine and chemokine expression with L-Arg treatment. Genomic analysis by microarray demonstrated that DSS-treated mice supplemented with L-Arg clustered more closely with mice not exposed to DSS than to those receiving DSS alone, and revealed that multiple genes that were upregulated or downregulated with DSS alone exhibited normalization of expression with L-Arg supplementation. Additionally, L-Arg treatment of mice with DSS colitis resulted in increased ex vivo migration of colonic epithelial cells, suggestive of increased capacity for wound repair. Because CAT2 induction was sustained during L-Arg treatment and inducible nitric oxide (NO) synthase (iNOS) requires uptake of L-Arg for generation of NO, we tested the effect of L-Arg in iNOS(-/-) mice and found that its benefits in DSS colitis were eliminated. These preclinical studies indicate that L-Arg supplementation could be a potential therapy for IBD, and that one mechanism of action may be functional enhancement of iNOS activity.


Subject(s)
Arginine/pharmacology , Arginine/pharmacokinetics , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Dextran Sulfate/toxicity , Gene Expression Regulation/drug effects , Amino Acids/blood , Amino Acids/metabolism , Animals , Arginine/therapeutic use , Blotting, Western , Cationic Amino Acid Transporter 2/metabolism , Cell Movement/drug effects , Chemokines/metabolism , Colitis, Ulcerative/pathology , Cytokines/metabolism , DNA Primers/genetics , Dietary Supplements , Epithelial Cells/drug effects , Gene Expression Regulation/physiology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Neutrophils/drug effects , Nitric Oxide Synthase Type II/genetics , Organ Size/drug effects , Permeability/drug effects , Weight Loss/drug effects
20.
Am J Physiol Gastrointest Liver Physiol ; 302(9): G1061-73, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22361732

ABSTRACT

Restoration of the colonic epithelial barrier is an important response during colitis. L-arginine (L-Arg) is a semiessential amino acid that reduces murine colitis induced by Citrobacter rodentium. Cationic amino acid transporter (CAT) proteins increase L-Arg uptake into cells. L-Arg is utilized to produce nitric oxide (NO), by inducible NO synthase (iNOS), or L-ornithine (L-Orn) by arginase (Arg) enzymes. The latter is followed by generation of polyamines by ornithine decarboxylase (ODC) and L-proline (L-Pro) by ornithine aminotransferase (OAT). We show that L-Arg enhanced epithelial restitution in conditionally immortalized young adult mouse colon (YAMC) cells in a wound repair model, and in isolated mouse colonic epithelial cells (CECs), using a cell migration assay. Restitution was impaired by C. rodentium. Wounding induced CAT2, and inhibition of L-Arg uptake by the competitive inhibitor L-lysine (L-Lys) or by CAT2 shRNA, but not CAT1 shRNA, decreased restitution. Migration was impaired in CECs treated with L-Lys or from CAT2(-/-) mice. Wounding increased Arg1 expression, and inhibition of arginase with S-(2-boronoethyl)-L-cysteine (BEC) or Arg1 shRNA inhibited restitution in YAMC cells; cell migration in CECs was also impaired by BEC. Inhibition of ODC or iNOS did not alter restitution. L-Orn or L-Pro restored restitution in cells treated with BEC or Arg1 shRNA, whereas the polyamine putrescine had no benefit. Wounding increased OAT levels, OAT shRNA inhibited restitution, and L-Pro restored restitution in cells with OAT knockdown. Uptake of L-Arg, and its metabolism by Arg1 to L-Orn and conversion to L-Pro by OAT is essential for colonic epithelial wound repair.


Subject(s)
Arginine/pharmacokinetics , Cationic Amino Acid Transporter 2/metabolism , Colitis/drug therapy , Colitis/metabolism , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Animals , Arginine/administration & dosage , Colon , Epithelial Cells/drug effects , Intestinal Mucosa/drug effects , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...