Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Am J Physiol Gastrointest Liver Physiol ; 319(1): G63-G73, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32538139

ABSTRACT

Hyaluronic acid (HA), a glycosaminoglycan in the extracellular matrix, binds to CD44 and Toll-like receptor 4 (TLR4). We previously demonstrated that both CD44 and TLR4, but predominately TLR4, mediated HA stimulation of Lgr5+ stem cell proliferation, crypt fission, and intestinal growth in postnatal mice. Here we address the questions of which cell type expresses the relevant TLR4 in driving intestinal growth and what are the downstream events from TLR4 activation. Studies were done in 14-day-old mice: wild type (WT), mice deficient in cyclooxygenase 2 (COX2), mice deficient in myeloid cell TLR4, and mice deficient in epithelial cell epidermal growth factor receptor (EGFR). Biological end points included crypt fission and Lgr5 cell proliferation. In WT mice, treatment with NS-398 (a COX2 inhibitor), clodronate (a macrophage-depleting agent), or tyrphostin (an EGFR inhibitor) resulted in 30% reductions in crypt fission and Lgr5+ stem cell proliferation compared with control mice. Mice deficient in COX2 or myeloid TLR4 or epithelial cell EGFR all had 30% reductions in crypt fission and Lgr5+ stem cell proliferation compared with WT mice. Administration of dimethyl PGE2, a stable PGE2 analog, increased crypt fission and Lgr5+ stem cell proliferation. Administration of dimethyl PGE2 reversed the effects of NS-398, clodronate, COX2 deficiency, and myeloid TLR4 deficiency but had no effect on mice treated with tyrphostin or mice deficient in epithelial cell EGFR. We conclude that, in postnatal mice, ~30% of intestinal growth as manifested by crypt fission and Lgr5+ stem cell proliferation is driven by a novel pathway: Extracellular HA binds TLR4 on pericryptal macrophages, inducing the production of PGE2 through COX2. PGE2 transactivates EGFR in Lgr5+ epithelial stem cells, resulting in Lgr5+ stem cell proliferation and crypt fission.NEW & NOTEWORTHY This study, in newborn mice, describes a novel molecular pathway regulating Lgr5+ epithelial stem cell proliferation and normal intestinal elongation, as assessed by crypt fission. In this pathway, endogenous extracellular hyaluronic acid binds to Toll-like receptor 4 on pericryptal macrophages releasing PGE2 which binds to epidermal growth factor receptor on Lgr5+ stem cells resulting in proliferation. Lgr5+ stem cell proliferation leads to crypt fission and intestinal elongation. The demonstration that normal growth requires microbial-independent Toll-like receptor activation is novel.


Subject(s)
Dinoprostone/metabolism , ErbB Receptors/drug effects , Hyaluronic Acid/pharmacology , Toll-Like Receptor 4/drug effects , Animals , Cell Proliferation/drug effects , Cyclooxygenase 2/metabolism , ErbB Receptors/metabolism , Hyaluronic Acid/antagonists & inhibitors , Intestines/drug effects , Mice, Knockout , Toll-Like Receptor 4/metabolism , Transcriptional Activation/drug effects
2.
Front Immunol ; 11: 617510, 2020.
Article in English | MEDLINE | ID: mdl-33552081

ABSTRACT

TLRs, key components of the innate immune system, recognize microbial molecules. However, TLRs also recognize some nonmicrobial molecules. In particular, TLR2 and TLR4 recognize hyaluronic acid, a glycosaminoglycan in the extracellular matrix. In neonatal mice endogenous hyaluronic acid binding to TLR4 drives normal intestinal growth. Hyaluronic acid binding to TLR4 in pericryptal macrophages results in cyclooxygenase2- dependent PGE2 production, which transactivates EGFR in LGR5+ crypt epithelial stem cells leading to increased proliferation. The expanded population of LGR5+ stem cells leads to crypt fission and lengthening of the intestine and colon. Blocking this pathway at any point (TLR4 activation, PGE2 production, EGFR transactivation) results in diminished intestinal and colonic growth. A similar pathway leads to epithelial proliferation in wound repair. The repair phase of dextran sodium sulfate colitis is marked by increased epithelial proliferation. In this model, TLR2 and TLR4 in pericryptal macrophages are activated by microbial products or by host hyaluronic acid, resulting in production of CXCL12, a chemokine. CXCL12 induces the migration of cyclooxygenase2-expressing mesenchymal stem cells from the lamina propria of the upper colonic crypts to a site adjacent to LGR5+ epithelial stem cells. PGE2 released by these mesenchymal stem cells transactivates EGFR in LGR5+ epithelial stem cells leading to increased proliferation. Several TLR2 and TLR4 agonists, including hyaluronic acid, are radioprotective in the intestine through the inhibition of radiation-induced apoptosis in LGR5+ epithelial stem cells. Administration of exogenous TLR2 or TLR4 agonists activates TLR2/TLR4 on pericryptal macrophages inducing CXCL12 production with migration of cyclooxygenase2-expressing mesenchymal stem cells from the lamina propria of the villi to a site adjacent to LGR5+ epithelial stem cells. PGE2 produced by these mesenchymal stem cells, blocks radiation-induced apoptosis in LGR5+ epithelial stem cells by an EGFR mediated pathway.


Subject(s)
Intestines/growth & development , Radiation Effects , Wound Healing/physiology , Animals , Cell Proliferation/physiology , Humans , Intestinal Mucosa/metabolism , Toll-Like Receptors/metabolism
3.
Mol Cancer Ther ; 18(12): 2446-2456, 2019 12.
Article in English | MEDLINE | ID: mdl-31484704

ABSTRACT

Hyaluronic acid (HA), a constituent of the extracellular matrix, promotes colorectal cancer growth. CD44 is a relevant HA receptor in this context. However, HA is also a ligand for TLR4, a receptor of significance in colorectal cancer. In this study, we examine the relative contribution of HA interactions with CD44 and TLR4 in colon tumorigenesis. Colorectal cancer models included ApcMin/+ mice, azoxymethane/dextran sodium sulfate (AOM-DSS), and CT26 tumor isografts. We used knockout mice and CT26 colorectal cancer cells with CRISPR knockdown of CD44 and TLR4. HA activity was modulated by PEP1 (a 12-mer peptide that blocks HA from binding its receptors), hyaluronidase (which promotes HA degradation), or 4-MU (HA synthesis inhibitor). Blockade of HA binding via PEP1 decreased growth in all colorectal cancer models and in cell culture. The effects were significant in WT and with CD44 deletion, but not with TLR4 deletion. In the AOM-DSS model, mice deficient in CD44 or TLR4 had fewer tumors. CD44- and TLR4-deficient CT26 isografts grew more slowly, exhibiting decreased tumor cell proliferation and increased apoptosis. In vitro, endogenous HA blocked LPS binding to TLR4 suggesting that HA is a relevant TLR4 ligand in colon cancer. Finally, PEP1 enhanced tumor radiation sensitivity in the isograft model. Together, these results indicate that HA binding to TLR4, as well as CD44, plays a key role in colon tumorigenesis. These findings also raise the possibility that an agent that blocks HA binding, such as PEP1, may be useful as an adjuvant therapy in colon cancer.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Hyaluronic Acid/therapeutic use , Toll-Like Receptor 4/drug effects , Animals , Apoptosis , Cell Line, Tumor , Colonic Neoplasms/pathology , Humans , Hyaluronic Acid/pharmacology , Mice
4.
Gut ; 68(6): 1003-1013, 2019 06.
Article in English | MEDLINE | ID: mdl-29934438

ABSTRACT

OBJECTIVE: Lactobacillus rhamnosus GG (LGG), a probiotic, given by gavage is radioprotective of the mouse intestine. LGG-induced radioprotection is toll-like receptor 2 (TLR2) and cyclooxygenase-2 (COX-2)-dependent and is associated with the migration of COX-2+mesenchymal stem cells (MSCs) from the lamina propria of the villus to the lamina propria near the crypt epithelial stem cells. Our goals were to define the mechanism of LGG radioprotection including identification of the TLR2 agonist, and the mechanism of the MSC migration and to determine the safety and efficacy of this approach in models relevant to clinical radiation therapy. DESIGN: Intestinal radioprotection was modelled in vitro with cell lines and enteroids as well as in vivo by assaying clinical outcomes and crypt survival. Fractionated abdominal and single dose radiation were used along with syngeneic CT26 colon tumour grafts to assess tumour radioprotection. RESULTS: LGG with a mutation in the processing of lipoteichoic acid (LTA), a TLR2 agonist, was not radioprotective, while LTA agonist and native LGG were. An agonist of CXCR4 blocked LGG-induced MSC migration and LGG-induced radioprotection. LGG given by gavage induced expression of CXCL12, a CXCR4 agonist, in pericryptal macrophages and depletion of macrophages by clodronate liposomes blocked LGG-induced MSC migration and radioprotection. LTA effectively protected the normal intestinal crypt, but not tumours in fractionated radiation regimens. CONCLUSIONS: LGG acts as a 'time-release capsule' releasing radioprotective LTA. LTA then primes the epithelial stem cell niche to protect epithelial stem cells by triggering a multicellular, adaptive immune signalling cascade involving macrophages and PGE2 secreting MSCs. TRIAL REGISTRATION NUMBER: NCT01790035; Pre-results.


Subject(s)
Intestinal Mucosa/metabolism , Lacticaseibacillus rhamnosus , Lipopolysaccharides/metabolism , Probiotics/pharmacology , Radiation Injuries/prevention & control , Teichoic Acids/metabolism , Animals , Cell Movement/radiation effects , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Intestinal Mucosa/drug effects , Intestinal Mucosa/radiation effects , Macrophage Activation/radiation effects , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Radiation-Protective Agents , Reference Values , Sensitivity and Specificity
5.
Immunity ; 49(1): 107-119.e4, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29958798

ABSTRACT

Intestinal macrophages are critical for gastrointestinal (GI) homeostasis, but our understanding of their role in regulating intestinal motility is incomplete. Here, we report that CX3C chemokine receptor 1-expressing muscularis macrophages (MMs) were required to maintain normal GI motility. MMs expressed the transient receptor potential vanilloid 4 (TRPV4) channel, which senses thermal, mechanical, and chemical cues. Selective pharmacologic inhibition of TRPV4 or conditional deletion of TRPV4 from macrophages decreased intestinal motility and was sufficient to reverse the GI hypermotility that is associated with chemotherapy treatment. Mechanistically, stimulation of MMs via TRPV4 promoted the release of prostaglandin E2 and elicited colon contraction in a paracrine manner via prostaglandin E receptor signaling in intestinal smooth muscle cells without input from the enteric nervous system. Collectively, our data identify TRPV4-expressing MMs as an essential component required for maintaining normal GI motility and provide potential drug targets for GI motility disorders.


Subject(s)
Colon/physiology , Gastrointestinal Motility , Macrophages/metabolism , Myocytes, Smooth Muscle/metabolism , Signal Transduction , TRPV Cation Channels/metabolism , Animals , CX3C Chemokine Receptor 1/metabolism , Colon/physiopathology , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/metabolism , Dinoprostone/analysis , Dinoprostone/metabolism , Female , Gastric Mucosa/cytology , Gene Expression , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mice, Knockout , Muscle Contraction , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics
6.
Dev Biol ; 409(2): 473-88, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26586201

ABSTRACT

Hirschsprung Disease (HSCR) is a potentially deadly birth defect characterized by the absence of the enteric nervous system (ENS) in distal bowel. Although HSCR has clear genetic causes, no HSCR-associated mutation is 100% penetrant, suggesting gene-gene and gene-environment interactions determine HSCR occurrence. To test the hypothesis that certain medicines might alter HSCR risk we treated zebrafish with medications commonly used during early human pregnancy and discovered that ibuprofen caused HSCR-like absence of enteric neurons in distal bowel. Using fetal CF-1 mouse gut slice cultures, we found that ibuprofen treated enteric neural crest-derived cells (ENCDC) had reduced migration, fewer lamellipodia and lower levels of active RAC1/CDC42. Additionally, inhibiting ROCK, a RHOA effector and known RAC1 antagonist, reversed ibuprofen effects on migrating mouse ENCDC in culture. Ibuprofen also inhibited colonization of Ret+/- mouse bowel by ENCDC in vivo and dramatically reduced bowel colonization by chick ENCDC in culture. Interestingly, ibuprofen did not affect ENCDC migration until after at least three hours of exposure. Furthermore, mice deficient in Ptgs1 (COX 1) and Ptgs2 (COX 2) had normal bowel colonization by ENCDC and normal ENCDC migration in vitro suggesting COX-independent effects. Consistent with selective and strain specific effects on ENCDC, ibuprofen did not affect migration of gut mesenchymal cells, NIH3T3, or WT C57BL/6 ENCDC, and did not affect dorsal root ganglion cell precursor migration in zebrafish. Thus, ibuprofen inhibits ENCDC migration in vitro and bowel colonization by ENCDC in vivo in zebrafish, mouse and chick, but there are cell type and strain specific responses. These data raise concern that ibuprofen may increase Hirschsprung disease risk in some genetically susceptible children.


Subject(s)
Cell Movement/drug effects , Enteric Nervous System/cytology , Ibuprofen/pharmacology , Intestines/cytology , Neural Stem Cells/cytology , Actin Cytoskeleton/metabolism , Animals , Caspase 3/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chickens , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/metabolism , Enzyme Activation/drug effects , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mesoderm/cytology , Mice , Models, Biological , NIH 3T3 Cells , Neural Stem Cells/drug effects , Neurons/cytology , Neurons/drug effects , Organ Culture Techniques , PPAR gamma/metabolism , Pseudopodia/drug effects , Pseudopodia/metabolism , Zebrafish , rac1 GTP-Binding Protein/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
7.
Am J Physiol Gastrointest Liver Physiol ; 309(11): G874-87, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26505972

ABSTRACT

Hyaluronic acid, a glycosaminoglycan in the extracellular matrix, binds to CD44 and Toll-like receptor 4 (TLR4). We previously addressed the role of hyaluronic acid in small intestinal and colonic growth in mice. We addressed the role of exogenous hyaluronic acid by giving hyaluronic acid intraperitoneally and the role of endogenous hyaluronic acid by giving PEP-1, a peptide that blocks hyaluronic acid binding to its receptors. Exogenous hyaluronic acid increased epithelial proliferation but had no effect on intestinal length. PEP-1 resulted in a shortened small intestine and colon and diminished epithelial proliferation. In the current study, we sought to determine whether the effects of hyaluronic acid on growth were mediated by signaling through CD44 or TLR4 by giving exogenous hyaluronic acid or PEP-1 twice a week from 3-8 wk of age to wild-type, CD44(-/-), and TLR4(-/-) mice. These studies demonstrated that signaling through both CD44 and TLR4 were important in mediating the effects of hyaluronic acid on growth in the small intestine and colon. Extending our studies to early postnatal life, we assessed the effects of exogenous hyaluronic acid and PEP-1 on Lgr5(+) stem cell proliferation and crypt fission. Administration of PEP-1 to Lgr5(+) reporter mice from postnatal day 7 to day 14 decreased Lgr5(+) cell proliferation and decreased crypt fission. These studies indicate that endogenous hyaluronic acid increases Lgr5(+) stem cell proliferation, crypt fission, and intestinal lengthening and that these effects are dependent on signaling through CD44 and TLR4.


Subject(s)
Cell Proliferation , Colon/metabolism , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Receptors, G-Protein-Coupled/metabolism , Stem Cells/drug effects , Toll-Like Receptor 4/metabolism , Animals , Biomarkers/metabolism , Cell Proliferation/drug effects , Colon/drug effects , Colon/growth & development , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Genotype , Hyaluronan Receptors/genetics , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/antagonists & inhibitors , Injections, Intraperitoneal , Intestinal Mucosa/drug effects , Intestinal Mucosa/growth & development , Intestine, Small/drug effects , Intestine, Small/growth & development , Male , Mice, Inbred C57BL , Mice, Knockout , Peptides/pharmacology , Phenotype , Signal Transduction/drug effects , Stem Cells/metabolism , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
8.
Curr Opin Support Palliat Care ; 9(2): 157-62, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25872116

ABSTRACT

PURPOSE OF REVIEW: There is currently an unmet need for agents that can prevent the gastrointestinal toxicity (mucositis and enteritis) associated with chemotherapy and radiation therapy of abdominal and pelvic cancers. Herein we provide an overview of how manipulation of the gut microbiota by probiotic administration affects these gastrointestinal symptoms. We focus this review on published human trials and also provide suggestions on how the field can move forward. RECENT FINDINGS: Several clinical trials of varying design, patient populations and probiotic products have been reported. Lactobacillus probiotics of adequate dosage demonstrate a potential to reduce gastrointestinal toxicity when administered prophylactically. Common study limitations prevent the widespread adoption of this practice at this point but are informative for rational design of future trials. SUMMARY: No single probiotic strain or product has emerged from human clinical trials for this indication. Further human studies are required to address limitations in the current literature. Preclinical model data should be used to inform the rational design of these new clinical trials to adequately address this important question.


Subject(s)
Antineoplastic Agents/adverse effects , Enteritis/prevention & control , Lactobacillus/immunology , Mucositis/prevention & control , Probiotics/therapeutic use , Radiotherapy/adverse effects , Abdominal Neoplasms/complications , Abdominal Neoplasms/drug therapy , Abdominal Neoplasms/radiotherapy , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic , Enteritis/etiology , Enteritis/microbiology , Enteritis/physiopathology , Humans , Lactobacillus/physiology , Microbiota/drug effects , Microbiota/radiation effects , Mucositis/etiology , Mucositis/microbiology , Mucositis/physiopathology , Pelvic Neoplasms/complications , Pelvic Neoplasms/drug therapy , Pelvic Neoplasms/radiotherapy
9.
Clin Gastroenterol Hepatol ; 13(6): 1197-200, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25460565

ABSTRACT

We performed a prospective study of patients with inflammatory bowel diseases to examine variations in treatment among medical centers. In a prospective cohort study of 1659 patients with Crohn's disease and 946 patients with ulcerative colitis seen at 7 high-volume referral centers, we collected data on demographics, disease characteristics, and medical and surgical treatments. We used logistic regression to determine differences in treatment among centers, controlling for potential confounders. We found significant variations among centers in the treatment of Crohn's disease with immunomodulators (odds ratio [OR], 3.34; 95% confidence interval [CI], 2.09-5.32) but not anti-tumor necrosis factor agents (OR, 1.64; 95% CI, 0.97-2.77). There was less variation in the treatment of ulcerative colitis; we found no difference in use of immunomodulators (OR, 1.83; 95% CI, 1.00-3.36) or anti-tumor necrosis factor therapy (OR, 0.81; 95% CI, 0.40-1.65). The development and implementation of evidence-based standards of care for inflammatory bowel disease may help reduce variation and improve outcomes.


Subject(s)
Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/surgery , Crohn Disease/drug therapy , Crohn Disease/surgery , Drug Therapy/methods , Drug Therapy/standards , Health Services Research , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Immunologic Factors/therapeutic use , Male , Middle Aged , Prospective Studies , United States , Young Adult
10.
Clin Cancer Res ; 21(6): 1466-76, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25538260

ABSTRACT

PURPOSE: Ulcerative colitis and colitis-associated colorectal cancer (CAC) is a serious health issue, but etiopathological factors remain unclear. Aldo-keto reductase 1B10 (AKR1B10) is specifically expressed in the colonic epithelium, but downregulated in colorectal cancer. This study was aimed to investigate the etiopathogenic role of AKR1B10 in ulcerative colitis and CAC. EXPERIMENTAL DESIGN: Ulcerative colitis and CAC biopsies (paraffin-embedded sections) and frozen tissues were collected to examine AKR1B10 expression. Aldo-keto reductase 1B8 (the ortholog of human AKR1B10) knockout (AKR1B8(-/-)) mice were produced to estimate its role in the susceptibility and severity of chronic colitis and associated dysplastic lesions, induced by dextran sulfate sodium (DSS) at a low dose (2%). Genome-wide exome sequencing was used to profile DNA damage in DSS-induced colitis and tumors. RESULTS: AKR1B10 expression was markedly diminished in over 90% of ulcerative colitis and CAC tissues. AKR1B8 deficiency led to reduced lipid synthesis from butyrate and diminished proliferation of colonic epithelial cells. The DSS-treated AKR1B8(-/-) mice demonstrated impaired injury repair of colonic epithelium and more severe bleeding, inflammation, and ulceration. These AKR1B8(-/-) mice had more severe oxidative stress and DNA damage, and dysplasias were more frequent and at a higher grade in the AKR1B8(-/-) mice than in wild-type mice. Palpable masses were seen in the AKR1B8(-/-) mice only, not in wild-type. CONCLUSIONS: AKR1B8 is a critical protein in the proliferation and injury repair of the colonic epithelium and in the pathogenesis of ulcerative colitis and CAC, being a new etiopathogenic factor of these diseases.


Subject(s)
Alcohol Oxidoreductases/genetics , Colitis, Ulcerative/pathology , Colon/pathology , Intestinal Mucosa/pathology , Oxidoreductases Acting on Aldehyde or Oxo Group Donors/metabolism , Alcohol Oxidoreductases/biosynthesis , Alcohol Oxidoreductases/metabolism , Aldo-Keto Reductases , Animals , Base Sequence , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Colitis, Ulcerative/chemically induced , Colorectal Neoplasms/pathology , DNA Damage/genetics , Dextran Sulfate , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/genetics , Oxidoreductases Acting on Aldehyde or Oxo Group Donors/biosynthesis , Oxidoreductases Acting on Aldehyde or Oxo Group Donors/genetics , Reactive Oxygen Species/metabolism , Sequence Analysis, DNA
11.
Curr Opin Gastroenterol ; 30(4): 347-51, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24837228

ABSTRACT

PURPOSE OF REVIEW: This review summarizes recent developments in the role of soluble mediators of inflammation, particularly arachidonic acid metabolites, in inflammatory bowel disease (IBD). RECENT FINDINGS: The role of prostaglandin E2 in immune regulation has been better defined. Prostaglandin E2 promotes not only immune tolerance and epithelial homeostasis but also the proinflammatory Th17 pathway. Prostaglandin D2 has been established as promoting the resolution of inflammation in the gastrointestinal mucosa. The 12-lipoxygenase product hepoxilin A3 mediates the migration of neutrophils from the mucosa into the lumen. SUMMARY: Recent studies of soluble mediators, especially arachidonic acid metabolites, have defined their proinflammatory and anti-inflammatory roles in IBD.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arachidonic Acid/immunology , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Th17 Cells/metabolism , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/metabolism , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Cell Movement , Dinoprostone/immunology , Humans , Inflammation/physiopathology , Inflammation/therapy , Inflammatory Bowel Diseases/physiopathology , Inflammatory Bowel Diseases/therapy , Neutrophils , Prostaglandin D2/immunology , Signal Transduction , Th17 Cells/immunology
13.
EBioMedicine ; 1(1): 46-57, 2014 Nov.
Article in English | MEDLINE | ID: mdl-26125048

ABSTRACT

The spread of multidrug-resistant microorganisms globally has created an urgent need for novel therapeutic strategies to combat urinary tract infections (UTIs). Immunomodulatory therapy may provide benefit, as treatment of mice with dexamethasone during acute UTI improved outcome by reducing the development of chronic cystitis, which predisposes to recurrent infection. Here we discovered soluble biomarkers engaged in myeloid cell development and chemotaxis that were predictive of future UTI recurrence when elevated in the sera of young women with UTI. Translation of these findings revealed that temperance of the neutrophil response early during UTI, and specifically disruption of bladder epithelial transmigration of neutrophils by inhibition of cyclooxygenase-2, protected mice against chronic and recurrent cystitis. Further, proteomics identified bladder epithelial remodeling consequent to chronic infection that enhances sensitivity to neutrophil damage. Thus, cyclooxygenase-2 expression during acute UTI is a critical molecular trigger determining disease outcome and drugs targeting cyclooxygenase-2 could prevent recurrent UTI.

14.
Gastroenterology ; 145(2): 416-25.e1-4, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23669411

ABSTRACT

BACKGROUND & AIMS: Indoleamine 2,3 dioxygenase-1 (IDO1) catabolizes tryptophan along the kynurenine pathway. Although IDO1 is expressed in inflamed and neoplastic epithelial cells of the colon, its role in colon tumorigenesis is not well understood. We used genetic and pharmacologic approaches to manipulate IDO1 activity in mice with colitis-associated cancer and human colon cancer cell lines. METHODS: C57Bl6 wild-type (control), IDO1-/-, Rag1-/-, and Rag1/IDO1 double-knockout mice were exposed to azoxymethane and dextran sodium sulfate to induce colitis and tumorigenesis. Colitis severity was assessed by measurements of disease activity, cytokine levels, and histologic analysis. In vitro experiments were conducted using HCT 116 and HT-29 human colon cancer cells. 1-methyl tryptophan and small interfering RNA were used to inhibit IDO1. Kynurenine pathway metabolites were used to simulate IDO1 activity. RESULTS: C57Bl6 mice given pharmacologic inhibitors of IDO1 and IDO1-/- mice had lower tumor burdens and reduced proliferation in the neoplastic epithelium after administration of dextran sodium sulfate and azoxymethane than control mice. These reductions also were observed in Rag1/IDO1 double-knockout mice compared with Rag1-/- mice (which lack mature adaptive immunity). In human colon cancer cells, blockade of IDO1 activity reduced nuclear and activated ß-catenin, transcription of its target genes (cyclin D1 and Axin2), and, ultimately, proliferation. Exogenous administration of IDO1 pathway metabolites kynurenine and quinolinic acid led to activation of ß-catenin and proliferation of human colon cancer cells, and increased tumor growth in mice. CONCLUSIONS: IDO1, which catabolizes tryptophan, promotes colitis-associated tumorigenesis in mice, independent of its ability to limit T-cell-mediated immune surveillance. The epithelial cell-autonomous survival advantage provided by IDO1 to colon epithelial cells indicate its potential as a therapeutic target.


Subject(s)
Colitis/physiopathology , Colonic Neoplasms/etiology , Epithelial Cells/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Intestinal Mucosa/physiopathology , beta Catenin/drug effects , Animals , Azoxymethane , Carcinogens , Cell Proliferation/drug effects , Colitis/metabolism , Colitis, Ulcerative , Colonic Neoplasms/chemically induced , Epithelial Cells/drug effects , Epithelial Cells/metabolism , HCT116 Cells , HT29 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Intestinal Mucosa/metabolism , Kynurenine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Quinolinic Acid/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , beta Catenin/physiology
15.
J Biol Chem ; 288(22): 16085-97, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23589310

ABSTRACT

The stem cell in the isthmus of gastric units continually replenishes the epithelium. Atrophy of acid-secreting parietal cells (PCs) frequently occurs during infection with Helicobacter pylori, predisposing patients to cancer. Atrophy causes increased proliferation of stem cells, yet little is known about how this process is regulated. Here we show that CD44 labels a population of small, undifferentiated cells in the gastric unit isthmus where stem cells are known to reside. Loss of CD44 in vivo results in decreased proliferation of the gastric epithelium. When we induce PC atrophy by Helicobacter infection or tamoxifen treatment, this CD44(+) population expands from the isthmus toward the base of the unit. CD44 blockade during PC atrophy abrogates the expansion. We find that CD44 binds STAT3, and inhibition of either CD44 or STAT3 signaling causes decreased proliferation. Atrophy-induced CD44 expansion depends on pERK, which labels isthmal cells in mice and humans. Our studies delineate an in vivo signaling pathway, ERK → CD44 → STAT3, that regulates normal and atrophy-induced gastric stem/progenitor-cell proliferation. We further show that we can intervene pharmacologically at each signaling step in vivo to modulate proliferation.


Subject(s)
Cell Proliferation , Helicobacter Infections/metabolism , Helicobacter pylori/metabolism , Hyaluronan Receptors/metabolism , Parietal Cells, Gastric/metabolism , Stem Cells/metabolism , Animals , Female , Helicobacter Infections/genetics , Helicobacter Infections/pathology , Humans , Hyaluronan Receptors/genetics , MAP Kinase Signaling System/genetics , Male , Metaplasia/genetics , Metaplasia/metabolism , Metaplasia/pathology , Mice , Parietal Cells, Gastric/pathology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Stem Cells/pathology
17.
FASEB J ; 27(3): 1191-202, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23233532

ABSTRACT

Genetic variants in the fatty acid (FA) translocase FAT/CD36 associate with abnormal postprandial lipids and influence risk for the metabolic syndrome. CD36 is abundant on apical enterocyte membranes in the proximal small intestine, where it facilitates FA uptake and FA-initiated signaling. We explored whether CD36 signaling influences FA-mediated secretion of cholecystokinin (CCK) and secretin, peptides released by enteroendocrine cells (EECs) in the duodenum/jejunum, which regulate events important for fat digestion and homeostasis. CD36 was immunodetected on apical membranes of secretin- and CCK-positive EECs and colocalized with cytosolic granules. Intragastric lipid administration to CD36 mice released less secretin (-60%) and CCK (-50%) compared with wild-type mice. Likewise, diminished secretin and CCK responses to FA were observed with CD36 intestinal segments in vitro, arguing against influence of alterations in fat absorption. Signaling mechanisms underlying peptide release were examined in STC-1 cells stably expressing human CD36 or a signaling-impaired mutant (CD36K/A). FA stimulation of cells expressing CD36 (vs. vector or CD36K/A) released more secretin (3.5- to 4-fold) and CCK (2- to 3-fold), generated more cAMP (2- to 2.5-fold), and enhanced protein kinase A activation. Protein kinase A inhibition (H-89) blunted secretin (80%) but not CCK release, which was reduced (50%) by blocking of calmodulin kinase II (KN-62). Coculture of STC-1 cells with Caco-2 cells stably expressing CD36 did not alter secretin or CCK release, consistent with a minimal effect of adjacent enterocytes. In summary, CD36 is a major mediator of FA-induced release of CCK and secretin. These peptides contribute to the role of CD36 in fat absorption and to its pleiotropic metabolic effects.


Subject(s)
CD36 Antigens/metabolism , Cholecystokinin/metabolism , Duodenum/metabolism , Enteroendocrine Cells/metabolism , Fatty Acids/metabolism , Jejunum/metabolism , Secretin/metabolism , Animals , CD36 Antigens/genetics , Caco-2 Cells , Cholecystokinin/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Duodenum/cytology , Enteroendocrine Cells/cytology , Enzyme Activation , Fatty Acids/genetics , Humans , Jejunum/cytology , Mice , Mice, Knockout , Secretin/genetics , Signal Transduction/physiology
18.
Am J Physiol Gastrointest Liver Physiol ; 303(3): G377-88, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22556141

ABSTRACT

Hyaluronic acid (HA), a component of the extracellular matrix, affects gastrointestinal epithelial proliferation in injury models, but its role in normal growth is unknown. We sought to determine the effects of exogenous HA on intestinal and colonic growth by intraperitoneal injection of HA twice a week into C57BL/6 mice from 3 to 8 wk of age. Similarly, to determine the effects of endogenous HA on intestinal and colonic growth, we administered PEP-1, a peptide that blocks the binding of HA to its receptors, on the same schedule. In mice treated with exogenous HA, villus height and crypt depth in the intestine, crypt depth in the colon, and epithelial proliferation in the intestine and colon were increased. In mice treated with PEP-1, intestinal and colonic length were markedly decreased and crypt depth and villus height in the intestine, crypt depth in the colon, and epithelial proliferation in the intestine and colon were decreased. Administration of HA was associated with increased levels of EGF (intestine) and IGF-I (colon), whereas administration of PEP-1 was associated with decreased levels of IGF-I (intestine) and epiregulin (colon). Exogenous HA increases intestinal and colonic epithelial proliferation, resulting in hyperplasia. Blocking the binding of endogenous HA to its receptors results in decreased intestinal and colonic length and a mucosal picture of hypoplasia, suggesting that endogenous HA contributes to the regulation of normal intestinal and colonic growth.


Subject(s)
Colon/growth & development , Hyaluronic Acid/pharmacology , Intestines/growth & development , Animals , Cell Proliferation/drug effects , Colon/drug effects , Epidermal Growth Factor/metabolism , Epiregulin , Epithelial Cells/cytology , Glucuronosyltransferase/biosynthesis , Hyaluronan Receptors/drug effects , Hyaluronan Synthases , Hyaluronoglucosaminidase/biosynthesis , Insulin-Like Growth Factor I/metabolism , Intestines/drug effects , Mice , Mice, Inbred C57BL , Peptides/pharmacology , RNA, Messenger/metabolism
19.
Inflamm Bowel Dis ; 18(7): 1214-20, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21823214

ABSTRACT

BACKGROUND: Indoleamine 2,3 dioxygenase-1 (IDO1) is a tryptophan catabolizing enzyme with immunotolerance-promoting functions. We sought to determine if increased gut expression of IDO1 in Crohn's disease (CD) would result in detectable changes in serum levels of tryptophan and the initial IDO1 pathway catabolite, kynurenine. METHODS: Individuals were prospectively enrolled through the Washington University Digestive Diseases Research Center. The Montreal Classification was used for disease phenotyping. Disease severity was categorized by the Physician's Global Assessment. Serum tryptophan and kynurenine were measured by high-pressure liquid chromatography. IDO1 immunohistochemical staining was performed on formalin-fixed tissue blocks. RESULTS: In all, 25 CD patients and 11 controls were enrolled. Eight CD patients had serum collected at two different timepoints and levels of disease activity compared. Strong IDO1 expression exists in both the lamina propria and epithelium during active CD compared to controls. Suppressed serum tryptophan levels and an elevated kynurenine/tryptophan (K/T) ratio were found in individuals with active CD as compared to those in remission or the control population. K/T ratios correlated positively with disease activity as well as with C-reactive protein and erythrocyte sedimentation rate. In the subgroup of CD patients with two serum measurements, tryptophan levels were elevated while kynurenine levels and the K/T ratio lowered as the disease activity lessened. CONCLUSIONS: IDO1 expression in CD is associated with lower serum tryptophan and an elevated K/T ratio. These levels may serve as a reasonable objective marker of gut mucosal immune activation and as a surrogate for CD activity.


Subject(s)
Biomarkers/blood , Crohn Disease/blood , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenine/blood , Tryptophan/blood , Adult , Case-Control Studies , Chromatography, High Pressure Liquid , Crohn Disease/diagnosis , Crohn Disease/enzymology , Female , Humans , Immunoenzyme Techniques , Male , Middle Aged , Prognosis , Prospective Studies , Young Adult
20.
Am J Physiol Gastrointest Liver Physiol ; 302(3): G309-16, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22038822

ABSTRACT

The intestinal epithelium is sensitive to radiation injury. Damage to the intestinal epithelium is dose limiting in radiation therapy of abdominal cancers. There is a need for agents that can be given before radiation therapy to protect the intestinal epithelium. C57BL6 mice were subjected to 12 Gy of total body radiation. Some mice received intraperitoneal hyaluronic acid (HA) before radiation. Mice were killed 6 h after radiation to assess radiation-induced apoptosis in the intestine; other mice were killed at 84 h to assess crypt survival. Total body radiation (12 Gy) resulted in increased expression of HA synthases and HA in the intestine and increased plasma HA (5-fold). Intraperitoneal injection of HA (30 mg/kg) before radiation resulted in a 1.8-fold increase in intestinal crypt survival and a decrease in radiation-induced apoptosis. The radioprotective effects of HA were not seen in Toll-like receptor 4 (TLR4)- or cyclooxygenase-2 (COX-2)-deficient mice. Intraperitoneal injection of HA induced a 1.5-fold increase in intestinal COX-2 expression, a 1.5-fold increase in intestinal PGE2, and the migration of COX-2-expressing mesenchymal stem cells from the lamina propria in the villi to the lamina propria near the crypt. We conclude that 1) radiation induces increased HA expression through inducing HA synthases, 2) intraperitoneal HA given before radiation reduces radiation-induced apoptosis and increases crypt survival, and 3) these radioprotective effects are mediated through TLR4, COX-2, and the migration of COX-2-expressing mesenchymal stem cells.


Subject(s)
Cyclooxygenase 2/metabolism , Hyaluronic Acid/pharmacology , Intestines/drug effects , Intestines/radiation effects , Radiation-Protective Agents/pharmacology , Toll-Like Receptor 4/metabolism , Animals , Antigens, CD/metabolism , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Movement/drug effects , Cyclooxygenase 2/genetics , Dinoprostone/metabolism , Gene Expression/genetics , Gene Expression/radiation effects , Glucuronosyltransferase/genetics , Hyaluronan Receptors/genetics , Hyaluronan Synthases , Hyaluronic Acid/antagonists & inhibitors , Hyaluronic Acid/blood , Hyaluronic Acid/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/radiation effects , Intestines/pathology , Jejunum/drug effects , Jejunum/metabolism , Jejunum/pathology , Jejunum/radiation effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Radiation-Protective Agents/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...