Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Biol Cell ; 26(12): 2252-62, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25904334

ABSTRACT

Intestinal epithelial cells (IECs) are exposed to profound fluctuations in oxygen tension and have evolved adaptive transcriptional responses to a low-oxygen environment. These adaptations are mediated primarily through the hypoxia-inducible factor (HIF) complex. Given the central role of the IEC in barrier function, we sought to determine whether HIF influenced epithelial tight junction (TJ) structure and function. Initial studies revealed that short hairpin RNA-mediated depletion of the HIF1ß in T84 cells resulted in profound defects in barrier and nonuniform, undulating TJ morphology. Global HIF1α chromatin immunoprecipitation (ChIP) analysis identified claudin-1 (CLDN1) as a prominent HIF target gene. Analysis of HIF1ß-deficient IEC revealed significantly reduced levels of CLDN1. Overexpression of CLDN1 in HIF1ß-deficient cells resulted in resolution of morphological abnormalities and restoration of barrier function. ChIP and site-directed mutagenesis revealed prominent hypoxia response elements in the CLDN1 promoter region. Subsequent in vivo analysis revealed the importance of HIF-mediated CLDN1 expression during experimental colitis. These results identify a critical link between HIF and specific tight junction function, providing important insight into mechanisms of HIF-regulated epithelial homeostasis.


Subject(s)
Claudin-1/genetics , Hypoxia-Inducible Factor 1/physiology , Intestinal Mucosa/physiology , Tight Junctions/physiology , Chromatin Immunoprecipitation , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Signal Transduction , Tight Junctions/metabolism , Transcriptional Activation
2.
Cell Host Microbe ; 17(5): 662-71, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25865369

ABSTRACT

Interactions between the microbiota and distal gut are fundamental determinants of human health. Such interactions are concentrated at the colonic mucosa and provide energy for the host epithelium through the production of the short-chain fatty acid butyrate. We sought to determine the role of epithelial butyrate metabolism in establishing the austere oxygenation profile of the distal gut. Bacteria-derived butyrate affects epithelial O2 consumption and results in stabilization of hypoxia-inducible factor (HIF), a transcription factor coordinating barrier protection. Antibiotic-mediated depletion of the microbiota reduces colonic butyrate and HIF expression, both of which are restored by butyrate supplementation. Additionally, germ-free mice exhibit diminished retention of O2-sensitive dyes and decreased stabilized HIF. Furthermore, the influences of butyrate are lost in cells lacking HIF, thus linking butyrate metabolism to stabilized HIF and barrier function. This work highlights a mechanism where host-microbe interactions augment barrier function in the distal gut.


Subject(s)
Bacteria/metabolism , Epithelial Cells/drug effects , Epithelial Cells/physiology , Fatty Acids, Volatile/metabolism , Gene Expression Regulation/drug effects , Hypoxia-Inducible Factor 1/biosynthesis , Animals , Cell Line , Epithelial Cells/metabolism , Humans , Mice , Oxygen Consumption
3.
Immunity ; 40(1): 66-77, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24412613

ABSTRACT

Acute intestinal inflammation involves early accumulation of neutrophils (PMNs) followed by either resolution or progression to chronic inflammation. Based on recent evidence that mucosal metabolism influences disease outcomes, we hypothesized that transmigrating PMNs influence the transcriptional profile of the surrounding mucosa. Microarray studies revealed a cohort of hypoxia-responsive genes regulated by PMN-epithelial crosstalk. Transmigrating PMNs rapidly depleted microenvironmental O2 sufficiently to stabilize intestinal epithelial cell hypoxia-inducible factor (HIF). By utilizing HIF reporter mice in an acute colitis model, we investigated the relative contribution of PMNs and the respiratory burst to "inflammatory hypoxia" in vivo. CGD mice, lacking a respiratory burst, developed accentuated colitis compared to control, with exaggerated PMN infiltration and diminished inflammatory hypoxia. Finally, pharmacological HIF stabilization within the mucosa protected CGD mice from severe colitis. In conclusion, transcriptional imprinting by infiltrating neutrophils modulates the host response to inflammation, via localized O2 depletion, resulting in microenvironmental hypoxia and effective inflammatory resolution.


Subject(s)
Colitis/immunology , Hypoxia/immunology , Mucous Membrane/metabolism , Neutrophils/pathology , Animals , Cell Communication , Cell Movement , Cells, Cultured , Cellular Microenvironment , Colitis/chemically induced , Colon/pathology , Disease Models, Animal , Hypoxia/chemically induced , Hypoxia-Inducible Factor 1/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Mucous Membrane/pathology , NADPH Oxidase 2 , NADPH Oxidases/genetics , Oxidative Stress , Oxygen/metabolism , Protein Stability/drug effects , Transendothelial and Transepithelial Migration
4.
J Immunol ; 192(3): 1267-76, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24367025

ABSTRACT

Cytokines secreted at sites of inflammation impact the onset, progression, and resolution of inflammation. In this article, we investigated potential proresolving mechanisms of IFN-γ in models of inflammatory bowel disease. Guided by initial microarray analysis, in vitro studies revealed that IFN-γ selectively induced the expression of IL-10R1 on intestinal epithelia. Further analysis revealed that IL-10R1 was expressed predominantly on the apical membrane of polarized epithelial cells. Receptor activation functionally induced canonical IL-10 target gene expression in epithelia, concomitant with enhanced barrier restitution. Furthermore, knockdown of IL-10R1 in intestinal epithelial cells results in impaired barrier function in vitro. Colonic tissue isolated from murine colitis revealed that levels of IL-10R1 and suppressor of cytokine signaling 3 were increased in the epithelium and coincided with increased tissue IFN-γ and IL-10 cytokines. In parallel, studies showed that treatment of mice with rIFN-γ was sufficient to drive expression of IL-10R1 in the colonic epithelium. Studies of dextran sodium sulfate colitis in intestinal epithelial-specific IL-10R1-null mice revealed a remarkable increase in disease susceptibility associated with increased intestinal permeability. Together, these results provide novel insight into the crucial and underappreciated role of epithelial IL-10 signaling in the maintenance and restitution of epithelial barrier and of the temporal regulation of these pathways by IFN-γ.


Subject(s)
Epithelial Cells/metabolism , Interferon-gamma/pharmacology , Interleukin-10 Receptor alpha Subunit/biosynthesis , Interleukin-10/physiology , Intestinal Mucosa/metabolism , Animals , Cell Line , Cell Polarity , Colitis/chemically induced , Colitis/metabolism , Cytokines/biosynthesis , Cytokines/genetics , Dextran Sulfate/toxicity , Dextrans/pharmacokinetics , Epithelial Cells/drug effects , Epithelial Cells/ultrastructure , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/pharmacokinetics , Gene Expression Regulation , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/physiology , Interleukin-10 Receptor alpha Subunit/genetics , Mice , Mice, Inbred C57BL , Permeability , Recombinant Proteins/pharmacology , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/biosynthesis , Suppressor of Cytokine Signaling Proteins/genetics
5.
Proc Natl Acad Sci U S A ; 110(49): 19820-5, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24248342

ABSTRACT

Mucosal surfaces of the lower gastrointestinal tract are subject to frequent, pronounced fluctuations in oxygen tension, particularly during inflammation. Adaptive responses to hypoxia are orchestrated largely by the hypoxia-inducible transcription factors (HIFs). As HIF-1α and HIF-2α are coexpressed in mucosal epithelia that constitute the barrier between the lumen and the underlying immune milieu, we sought to define the discrete contribution of HIF-1 and HIF-2 transactivation pathways to intestinal epithelial cell homeostasis. The present study identifies creatine kinases (CKs), key metabolic enzymes for rapid ATP generation via the phosphocreatine-creatine kinase (PCr/CK) system, as a unique gene family that is coordinately regulated by HIF. Cytosolic CKs are expressed in a HIF-2-dependent manner in vitro and localize to apical intestinal epithelial cell adherens junctions, where they are critical for junction assembly and epithelial integrity. Supplementation with dietary creatine markedly ameliorated both disease severity and inflammatory responses in colitis models. Further, enzymes of the PCr/CK metabolic shuttle demonstrate dysregulated mucosal expression in a subset of ulcerative colitis and Crohn disease patients. These findings establish a role for HIF-regulated CK in epithelial homeostasis and reveal a fundamental link between cellular bioenergetics and mucosal barrier.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Cell Hypoxia/physiology , Colitis/metabolism , Creatine Kinase/metabolism , Creatine/metabolism , Gene Expression Regulation, Enzymologic/physiology , Signal Transduction/physiology , Analysis of Variance , Blotting, Western , Chromatography, High Pressure Liquid , DNA Primers/genetics , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation, Enzymologic/genetics , Gene Knockdown Techniques , Humans , Immunoprecipitation , Polymerase Chain Reaction
6.
J Immunol ; 190(1): 392-400, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23209320

ABSTRACT

A deeper understanding of the mechanisms that control responses to inflammation is critical to the development of effective therapies. We sought to define the most proximal regulators of the Cullin (Cul)-RING ligases, which play a central role in the stabilization of NF-κB and hypoxia-inducible factor (HIF). In these studies, we identify the human deneddylase-1 (SENP8) as a key regulator of Cul neddylation response in vitro and in vivo. Using human microvascular endothelial cells (HMECs), we examined inflammatory responses to LPS or TNF-α by assessing Cul neddylation status, NF-κB and HIF-1α stabilization, and inflammatory cytokine secretion. HMECs with an intact neddylation pathway showed a time-dependent induction of Cul-1 neddylation, nuclear translocation of NF-κB, stabilization of HIF-1α, and increased NF-κB/HIF-α promoter activity in response to LPS. HMECs lacking SENP8 were unable to neddylate Cul-1 and subsequently were unable to activate NF-κB or HIF-1α. Pharmacological targeting of neddylation (MLN4924) significantly abrogated NF-κB responses, induced HIF-1α promoter activity, and reduced secretion of TNF-α-elicited proinflammatory cytokines. MLN4924 stabilized HIF and abrogated proinflammatory responses while maintaining anti-inflammatory IL-10 responses in vivo following LPS administration. These studies identify SENP8 as a proximal regulator of Cul neddylation and provide an important role for SENP8 in fine-tuning the inflammatory response. Moreover, our findings provide feasibility for therapeutic targeting of the Culs during inflammation.


Subject(s)
Cullin Proteins/physiology , Endopeptidases/physiology , Endothelium, Vascular/enzymology , Endothelium, Vascular/immunology , Inflammation Mediators/physiology , Ubiquitins/physiology , Cells, Cultured , Cullin Proteins/metabolism , Endopeptidases/deficiency , Endopeptidases/genetics , Endothelium, Vascular/cytology , Enzyme Precursors/metabolism , Enzyme Precursors/physiology , Human Umbilical Vein Endothelial Cells , Humans , Microcirculation/immunology , NEDD8 Protein , Ubiquitins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...