Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
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
2.
Proc Natl Acad Sci U S A ; 117(21): 11648-11657, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32398370

ABSTRACT

The intestinal mucosa exists in dynamic balance with trillions of luminal microbes. Disruption of the intestinal epithelial barrier, commonly observed in mucosal inflammation and diseases such as inflammatory bowel diseases (IBDs), is often associated with dysbiosis, particularly decreases in species producing short-chain fatty acids (SCFAs), such as butyrate. It remains unclear to what extent microbiota-derived factors contribute to the overall maintenance of intestinal homeostasis. Initial studies revealed that butyrate selectively promotes epithelial barrier function and wound healing. We aimed to define the specific mechanism(s) through which butyrate contributes to these epithelial responses. Guided by an unbiased profiling approach, we identified the dominant regulation of the actin-binding protein synaptopodin (SYNPO). Extensions of this work revealed a role for SYNPO in intestinal epithelial barrier function and wound healing. SYNPO was localized to the intestinal epithelial tight junction and within F-actin stress fibers where it is critical for barrier integrity and cell motility. Butyrate, but not other SCFAs, induced SYNPO in epithelial cell lines and murine colonic enteroids through mechanisms possibly involving histone deacetylase inhibition. Moreover, depletion of the microbiota abrogated expression of SYNPO in the mouse colon, which was rescued with butyrate repletion. Studies in Synpo-deficient mice demonstrated exacerbated disease susceptibility and increased intestinal permeability in a dextran sulfate sodium colitis model. These findings establish a critical role for the microbiota and their products, specifically butyrate, in the regulated expression of SYNPO for intestinal homeostasis and reveal a direct mechanistic link between microbiota-derived butyrate and barrier restoration.


Subject(s)
Butyrates/metabolism , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/metabolism , Microfilament Proteins , Animals , Cell Line , Homeostasis/physiology , Humans , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Tight Junctions/metabolism
3.
FASEB J ; 35(6): e21551, 2021 06.
Article in English | MEDLINE | ID: mdl-34042222

ABSTRACT

Intestinal epithelial cells (IEC) are crucial for maintaining proper digestion and overall homeostasis of the gut mucosa. IEC proliferation and differentiation are tightly regulated by well described pathways, however, relatively little is known about how cytokines shape these processes. Given that the anti-inflammatory cytokine interleukin (IL)-10 promotes intestinal barrier function, and insufficient IL-10 signaling increases susceptibility to intestinal diseases like inflammatory bowel disease, we hypothesized that IL-10 signaling modulates processes underlying IEC proliferation and differentiation. This was tested using in vivo and in vitro IEC-specific IL-10 receptor 1 (IL-10R1) depletion under homeostatic conditions. Our findings revealed that loss of IL-10R1 drove lineage commitment toward a dominant goblet cell phenotype while decreasing absorptive cell-related features. Diminished IL-10 signaling also significantly elevated IEC proliferation with relatively minor changes to apoptosis. Characterization of signaling pathways upstream of proliferation demonstrated a significant reduction in the Wnt inhibitor, DKK1, increased nuclear localization of ß-catenin, and increased transcripts of the proliferation marker, OLFM4, with IL-10R1 depletion. Phosphorylated STAT3 was nearly completely absent in IL-10R1 knockdown cells and may provide a mechanistic link between our observations and the regulation of these cellular processes. Our results demonstrate a novel role for IL-10 signaling in intestinal mucosal homeostasis by regulating proper balance of proliferation and IEC lineage fate.


Subject(s)
Cell Differentiation , Cell Proliferation , Epithelial Cells/pathology , Goblet Cells/pathology , Intestinal Mucosa/pathology , Receptors, Interleukin-10/physiology , Animals , Apoptosis , Epithelial Cells/metabolism , Female , Goblet Cells/metabolism , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
4.
Immunol Rev ; 273(1): 112-20, 2016 09.
Article in English | MEDLINE | ID: mdl-27558331

ABSTRACT

The interaction of neutrophils (PMNs) and epithelial cells are requisite lines of communication during mucosal inflammatory responses. Consequences of such interactions often determine endpoint organ function, and for this reason, much interest has developed around defining the constituents of the tissue microenvironment of inflammatory lesions. Physiologic in vitro and in vivo models have aided in the discovery of components that define the basic inflammatory machinery that mold the inflammatory tissue microenvironment. Here, we will review the recent literature related to the contribution of PMNs to molding of the tissue microenvironment, with an emphasis on the gastrointestinal (GI) tract. We focus on endogenous pathways for promoting tissue homeostasis and the molecular determinants of neutrophil-epithelial cell interactions during ongoing inflammation. These recent studies highlight the dynamic nature of these pathways and lend insight into the complexity of treating mucosal inflammation.


Subject(s)
Cellular Microenvironment , Epithelial Cells/physiology , Inflammation/immunology , Intestinal Mucosa/physiology , Neutrophils/physiology , Animals , Cell Communication , Cell Movement , Homeostasis , Humans
5.
Am J Pathol ; 188(5): 1183-1194, 2018 05.
Article in English | MEDLINE | ID: mdl-29454749

ABSTRACT

Interactions between the gut microbiota and the host are important for health, where dysbiosis has emerged as a likely component of mucosal disease. The specific constituents of the microbiota that contribute to mucosal disease are not well defined. The authors sought to define microbial components that regulate homeostasis within the intestinal mucosa. Using an unbiased, metabolomic profiling approach, a selective depletion of indole and indole-derived metabolites was identified in murine and human colitis. Indole-3-propionic acid (IPA) was selectively diminished in circulating serum from human subjects with active colitis, and IPA served as a biomarker of disease remission. Administration of indole metabolites showed prominent induction of IL-10R1 on cultured intestinal epithelia that was explained by activation of the aryl hydrocarbon receptor. Colonization of germ-free mice with wild-type Escherichia coli, but not E. coli mutants unable to generate indole, induced colonic epithelial IL-10R1. Moreover, oral administration of IPA significantly ameliorated disease in a chemically induced murine colitis model. This work defines a novel role of indole metabolites in anti-inflammatory pathways mediated by epithelial IL-10 signaling and identifies possible avenues for utilizing indoles as novel therapeutics in mucosal disease.


Subject(s)
Colitis/metabolism , Indoles/metabolism , Intestinal Mucosa/metabolism , Microbiota/physiology , Receptors, Interleukin-10/metabolism , Animals , Cell Line , Colitis/chemically induced , Dextran Sulfate , Disease Models, Animal , Homeostasis/physiology , Humans , Metabolomics , Mice
6.
J Biol Chem ; 291(22): 11800-8, 2016 May 27.
Article in English | MEDLINE | ID: mdl-27044749

ABSTRACT

Molecular oxygen and carbon dioxide are the primary gaseous substrate and product of oxidative metabolism, respectively. Hypoxia (low oxygen) and hypercapnia (high carbon dioxide) are co-incidental features of the tissue microenvironment in a range of pathophysiologic states, including acute and chronic respiratory diseases. The hypoxia-inducible factor (HIF) is the master regulator of the transcriptional response to hypoxia; however, little is known about the impact of hypercapnia on gene transcription. Because of the relationship between hypoxia and hypercapnia, we investigated the effect of hypercapnia on the HIF pathway. Hypercapnia suppressed HIF-α protein stability and HIF target gene expression both in mice and cultured cells in a manner that was at least in part independent of the canonical O2-dependent HIF degradation pathway. The suppressive effects of hypercapnia on HIF-α protein stability could be mimicked by reducing intracellular pH at a constant level of partial pressure of CO2 Bafilomycin A1, a specific inhibitor of vacuolar-type H(+)-ATPase that blocks lysosomal degradation, prevented the hypercapnic suppression of HIF-α protein. Based on these results, we hypothesize that hypercapnia counter-regulates activation of the HIF pathway by reducing intracellular pH and promoting lysosomal degradation of HIF-α subunits. Therefore, hypercapnia may play a key role in the pathophysiology of diseases where HIF is implicated.


Subject(s)
Carbon Dioxide/blood , Hypercapnia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/physiopathology , Oxygen/metabolism , Animals , Blotting, Western , Cells, Cultured , Female , HCT116 Cells , HeLa Cells , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
7.
Blood ; 126(17): 1972-3, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26494916

ABSTRACT

In this issue of Blood, Massena et al identify a novel CD49d+CXCR4highVEGFR1high population of neutrophils that specifically migrate to sites of hypoxia and enhance angiogenesis.


Subject(s)
Integrin alpha4/metabolism , Islets of Langerhans/metabolism , Muscle, Skeletal/metabolism , Neutrophils/metabolism , Receptors, CXCR4/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/physiology , Animals , Female , Humans , Male
8.
FASEB J ; 29(1): 208-15, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25326537

ABSTRACT

There is interest in understanding post-translational modifications of proteins in inflammatory disease. Neddylation is the conjugation of the molecule neural precursor cell expressed, developmentally down-regulated 8 (NEDD8) to promote protein stabilization. Cullins are a family of NEDD8 targets important in the stabilization and degradation of proteins, such as hypoxia-inducible factor (HIF; via Cullin-2). Here, we elucidate the role of human deneddylase-1 (DEN-1, also called SENP8) in inflammatory responses in vitro and in vivo and define conditions for targeting neddylation in models of mucosal inflammation. HIF provides protection in inflammatory models, so we examined the contribution of DEN-1 to HIF stabilization. Pharmacologic targeting of neddylation activity with MLN4924 (IC50, 4.7 nM) stabilized HIF-1α, activated HIF promoter activity by 2.5-fold, and induced HIF-target genes in human epithelial cells up to 5-fold. Knockdown of DEN-1 in human intestinal epithelial cells resulted in increased kinetics in barrier formation, decreased permeability, and enhanced barrier restitution by 2 ± 0.5-fold. Parallel studies in vivo revealed that MLN4924 abrogated disease severity in murine dextran sulfate sodium colitis, including weight loss, colon length, and histologic severity. We conclude that DEN-1 is a regulator of cullin neddylation and fine-tunes the inflammatory response in vitro and in vivo. Pharmacologic inhibition of cullin neddylation may provide a therapeutic opportunity in mucosal inflammatory disease.


Subject(s)
Cullin Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/prevention & control , Animals , Cell Line , Cullin Proteins/antagonists & inhibitors , Cyclopentanes/pharmacology , Disease Models, Animal , Endopeptidases/genetics , Endopeptidases/metabolism , Gene Knockdown Techniques , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Metabolic Networks and Pathways , Mice, Inbred C57BL , NEDD8 Protein , Protease Inhibitors/pharmacology , Protein Stability , Pyrimidines/pharmacology , Ubiquitins/metabolism
9.
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
10.
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
11.
FASEB J ; 28(1): 256-64, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24029533

ABSTRACT

Hypoxia has been widely implicated in many pathological conditions, including those associated with inflammation and tumorigenesis. A number of recent studies have implicated hypoxia in the control of vasculogenesis and permeability, the basis for which is not fully understood. Here we examine the transcriptional regulation of angiogenesis and permeability by hypoxia in endothelial cells. Guided by a global profiling approach in cultured endothelial cells, these studies revealed the selective induction of human gravin (protein kinase A anchoring protein 12) by hypoxia. Analysis of the cloned gravin promoter identified a functional hypoxia-responsive region including 2 binding sites for hypoxia-inducible factor (HIF). Site-directed mutagenesis identified the most distal HIF-binding site as essential for the induction of gravin by hypoxia. Further studies examining gravin gain and loss of function confirmed strong dependence of gravin in control of microvascular endothelial tube formation, wherein gravin functions as a "braking" system for angiogenesis. Additional studies in confluent endothelia revealed that gravin functionally couples to control endothelial barrier function in response to protein kinase A (PKA) agonists. Taken together, these results demonstrate transcriptional coordination of gravin by HIF-1α and amplified PKA-dependent endothelial responses. These findings provide an important link between hypoxia and metabolic conditions associated with inflammation and angiogenesis.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Cycle Proteins/metabolism , Hypoxia-Inducible Factor 1/metabolism , A Kinase Anchor Proteins/genetics , Cell Cycle Proteins/genetics , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Line , Humans , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mutagenesis, Site-Directed
12.
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
13.
Proc Natl Acad Sci U S A ; 109(41): E2784-93, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-22988108

ABSTRACT

Recent studies have demonstrated dramatic shifts in metabolic supply-and-demand ratios during inflammation, a process resulting in localized tissue hypoxia within inflammatory lesions ("inflammatory hypoxia"). As part of the adaptive immune response, T cells are recruited to sites of inflammatory hypoxia. Given the profound effects of hypoxia on gene regulation, we hypothesized that T-cell differentiation is controlled by hypoxia. To pursue this hypothesis, we analyzed the transcriptional consequences of ambient hypoxia (1% oxygen) on a broad panel of T-cell differentiation factors. Surprisingly, these studies revealed selective, robust induction of FoxP3, a key transcriptional regulator for regulatory T cells (Tregs). Studies of promoter binding or loss- and gain-of-function implicated hypoxia-inducible factor (HIF)-1α in inducing FoxP3. Similarly, hypoxia enhanced Treg abundance in vitro and in vivo. Finally, Treg-intrinsic HIF-1α was required for optimal Treg function and Hif1a-deficient Tregs failed to control T-cell-mediated colitis. These studies demonstrate that hypoxia is an intrinsic molecular cue that promotes FoxP3 expression, in turn eliciting potent anti-inflammatory mechanisms to limit tissue damage in conditions of reduced oxygen availability.


Subject(s)
Forkhead Transcription Factors/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia , Inflammation/genetics , Intestinal Mucosa/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Hypoxia , Cell Proliferation , Cells, Cultured , Colitis/genetics , Colitis/metabolism , Female , Flow Cytometry , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/metabolism , Interleukin-1/pharmacology , Intestinal Mucosa/pathology , Jurkat Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta/pharmacology
14.
J Immunol ; 187(7): 3475-81, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21934099

ABSTRACT

Mucosal surfaces function as selectively permeable barriers between the host and the outside world. Given their close proximity to microbial Ags, mucosal surfaces have evolved sophisticated mechanisms for maintaining homeostasis and preventing excessive acute inflammatory reactions. The role attributed to epithelial cells was historically limited to serving as a selective barrier; in recent years, numerous findings implicate an active role of the epithelium with proresolving mediators in the maintenance of immunological equilibrium. In this brief review, we highlight new evidence that the epithelium actively contributes to coordination and resolution of inflammation, principally through the generation of anti-inflammatory and proresolution lipid mediators. These autacoids, derived from ω-6 and ω-3 polyunsaturated fatty acids, are implicated in the initiation, progression, and resolution of acute inflammation and display specific, epithelial-directed actions focused on mucosal homeostasis. We also summarize present knowledge of mechanisms for resolution via regulation of epithelial-derived antimicrobial peptides in response to proresolving lipid mediators.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Autacoids/immunology , Immunity, Mucosal/immunology , Inflammation/immunology , Animals , Humans , Inflammation/microbiology
15.
J Immunol ; 186(3): 1790-8, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21199896

ABSTRACT

Numerous studies have revealed that hypoxia and inflammation occur coincidentally in mucosal disorders, such as inflammatory bowel disease. During inflammation, epithelial-expressed hypoxia-inducible factor (HIF) serves an endogenously protective function. In this study, we sought to explore how mucosal immune responses influence HIF-dependent end points. Guided by a screen of relevant inflammatory mediators, we identified IFN-γ as a potent repressor of HIF-dependent transcription in human intestinal epithelial cells. Analysis of HIF levels revealed that HIF-1ß, but not HIF-1α, is selectively repressed by IFN-γ in a JAK-dependent manner. Cloning and functional analysis of the HIF-1ß promoter identified a prominent region for IFN-γ-dependent repression. Further studies revealed that colonic IFN-γ and HIF-1ß levels were inversely correlated in a murine colitis model. Taken together, these studies demonstrated that intestinal epithelial HIF is attenuated by IFN-γ through transcriptional repression of HIF-1ß. These observations are relevant to the pathophysiology of colitis (i.e., that loss of HIF signaling during active inflammation may exacerbate disease pathogenesis).


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/antagonists & inhibitors , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Colitis/immunology , Interferon-gamma/physiology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Repressor Proteins/physiology , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/physiology , Caco-2 Cells , Cell Line, Tumor , Cells, Cultured , Cloning, Molecular , Colitis/enzymology , Colitis/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/physiology , Intestinal Mucosa/enzymology , Mice , Mice, Inbred C57BL , Procollagen-Proline Dioxygenase/physiology , Signal Transduction/immunology
16.
J Immunol ; 186(11): 6505-14, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21515785

ABSTRACT

Tissues of the mucosa are lined by an epithelium that provides barrier and transport functions. It is now appreciated that inflammatory responses in inflammatory bowel diseases are accompanied by striking shifts in tissue metabolism. In this paper, we examined global metabolic consequences of mucosal inflammation using both in vitro and in vivo models of disease. Initial analysis of the metabolic signature elicited by inflammation in epithelial models and in colonic tissue isolated from murine colitis demonstrated that levels of specific metabolites associated with cellular methylation reactions are significantly altered by model inflammatory systems. Furthermore, expression of enzymes central to all cellular methylation, S-adenosylmethionine synthetase and S-adenosylhomocysteine hydrolase, are increased in response to inflammation. Subsequent studies showed that DNA methylation is substantially increased during inflammation and that epithelial NF-κB activity is significantly inhibited following treatment with a reversible S-adenosylhomocysteine hydrolase inhibitor, DZ2002. Finally, these studies demonstrated that inhibition of cellular methylation in a murine model of colitis results in disease exacerbation while folate supplementation to promote methylation partially ameliorates the severity of murine colitis. Taken together, these results identify a global change in methylation, which during inflammation, translates to an overall protective role in mucosal epithelia.


Subject(s)
Colitis/metabolism , Inflammation/metabolism , Intestinal Mucosa/metabolism , Metabolomics/methods , Adenine/analogs & derivatives , Adenine/pharmacology , Adenosylhomocysteinase/genetics , Adenosylhomocysteinase/metabolism , Animals , Blotting, Western , Butyrates/pharmacology , Cell Line, Tumor , Colitis/genetics , Colon/drug effects , Colon/metabolism , Colon/pathology , DNA Methylation/drug effects , Dextran Sulfate/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Profiling/methods , HeLa Cells , Humans , Inflammation/genetics , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Intestinal Mucosa/pathology , Magnetic Resonance Spectroscopy , Methionine Adenosyltransferase/genetics , Methionine Adenosyltransferase/metabolism , Methylation/drug effects , Mice , Mice, Inbred C57BL , Mucositis/genetics , Mucositis/metabolism , NF-kappa B/metabolism , Reverse Transcriptase Polymerase Chain Reaction
17.
Proc Natl Acad Sci U S A ; 107(32): 14298-303, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20660763

ABSTRACT

Resolvin-E1 (RvE1) has been demonstrated to promote inflammatory resolution in numerous disease models. Given the importance of epithelial cells to coordination of mucosal inflammation, we hypothesized that RvE1 elicits an epithelial resolution signature. Initial studies revealed that the RvE1-receptor (ChemR23) is expressed on intestinal epithelial cells (IECs) and that microarray profiling of cells exposed to RvE1 revealed regulation of inflammatory response gene expression. Notably, RvE1 induced intestinal alkaline phosphatase (ALPI) expression and significantly enhanced epithelial ALPI enzyme activity. One role recently attributed to ALPI is the detoxification of bacterial LPS. In our studies, RvE1-exposed epithelia detoxified LPS (assessed by attenuation of NF-kappaB signaling). Furthermore, in epithelial-bacterial interaction assays, we determined that ALPI retarded the growth of Escherichia coli. To define these features in vivo, we used a murine dextran sulfate sodium (DSS) model of colitis. Compared with vehicle controls, administration of RvE1 resulted in significant improvement of disease activity indices (e.g., body weight, colon length) concomitant with increased ALPI expression in the intestinal epithelium. Moreover, inhibition of ALPI activity resulted in increased severity of colitis in DSS-treated animals and partially abrogated the protective influence of RvE1. Together, these data implicate a previously unappreciated role for ALPI in RvE1-mediated inflammatory resolution.


Subject(s)
Alkaline Phosphatase/genetics , Eicosapentaenoic Acid/analogs & derivatives , Inflammation/prevention & control , Intestinal Mucosa/enzymology , Lipopolysaccharides/antagonists & inhibitors , Animals , Colitis/prevention & control , Eicosapentaenoic Acid/pharmacology , Epithelial Cells/chemistry , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/analysis , Transcriptional Activation
18.
mBio ; 14(1): e0312122, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36598189

ABSTRACT

Klebsiella pneumoniae is a leading cause of nosocomial and community acquired infections, making K. pneumoniae the pathogen that is associated with the second largest number of deaths attributed to any antibiotic resistant infection. K. pneumoniae colonizes the nasopharynx and the gastrointestinal tract in an asymptomatic manner without dissemination to other tissues. Importantly, gastrointestinal colonization is a requisite for infection. Our understanding of K. pneumoniae colonization is still based on interrogating mouse models in which animals are pretreated with antibiotics to disturb the colonization resistance imposed by the gut microbiome. In these models, infections disseminate to other tissues. Here, we report a murine model to allow for the study of the gastrointestinal colonization of K. pneumoniae without tissue dissemination. Hypervirulent and antibiotic resistant strains stably colonize the gastrointestinal tract of in an inbred mouse population without antibiotic treatment. The small intestine is the primary site of colonization and is followed by a transition to the colon over time, without dissemination to other tissues. Our model recapitulates the disease dynamics of the metastatic K. pneumoniae strains that are able to disseminate from the gastrointestinal tract to other sterile sites. Colonization is associated with mild to moderate histopathology, no significant inflammation, and no effect on the richness of the microbiome. Our model sums up the clinical scenario in which antibiotic treatment disturbs the colonization of K. pneumoniae and results in dissemination to other tissues. Finally, we establish that the capsule polysaccharide is necessary for the colonization of the large intestine, whereas the type VI secretion system contributes to colonization across the gastrointestinal tract. IMPORTANCE Klebsiella pneumoniae is one of the pathogens that is sweeping the world in the antibiotic resistance pandemic. Klebsiella colonizes the nasopharynx and the gut of healthy subjects in an asymptomatic manner, making gut colonization a requisite for infection. This makes it essential to understand the gastrointestinal carriage in preventing Klebsiella infections. Current research models rely on the perturbation of the gut microbiome by antibiotics, resulting in an invasive infection. Here, we report a new model of K. pneumoniae gut colonization that recapitulates key features of the asymptomatic human gastrointestinal tract colonization. In our model, there is no need to disturb the microbiota to achieve stable colonization, and there is no dissemination to other tissues. Our model sums up the clinical scenario in which antibiotic treatment triggers invasive infection. We envision that our model will be an excellent platform upon which to investigate factors enhancing colonization and invasive infections and to test therapeutics to eliminate Klebsiella asymptomatic colonization.


Subject(s)
Klebsiella Infections , Klebsiella pneumoniae , Humans , Animals , Mice , Gastrointestinal Tract/pathology , Anti-Bacterial Agents/pharmacology , Klebsiella Infections/epidemiology , Inflammation
19.
Discov Immunol ; 2(1): kyad018, 2023.
Article in English | MEDLINE | ID: mdl-38567056

ABSTRACT

Cross-talk between dendritic cells (DCs) and the intestinal epithelium is important in the decision to mount a protective immune response to a pathogen or to regulate potentially damaging responses to food antigens and the microbiota. Failures in this decision-making process contribute to the development of intestinal inflammation, making the molecular signals that pass between DCs and intestinal epithelial cells potential therapeutic targets. Until now, in vitro models with sufficient complexity to understand these interactions have been lacking. Here, we outline the development of a co-culture model of in vitro differentiated 'gut-like' DCs with small intestinal organoids (enteroids). Sequential exposure of murine bone marrow progenitors to Flt3L, granulocyte macrophage colony-stimulating factor (GM-CSF) and all-trans-retinoic acid (RA) resulted in the generation of a distinct population of conventional DCs expressing CD11b+SIRPα+CD103+/- (cDC2) exhibiting retinaldehyde dehydrogenase (RALDH) activity. These 'gut-like' DCs extended transepithelial dendrites across the intact epithelium of enteroids. 'Gut-like' DC in co-culture with enteroids can be utilized to define how epithelial cells and cDCs communicate in the intestine under a variety of different physiological conditions, including exposure to different nutrients, natural products, components of the microbiota, or pathogens. Surprisingly, we found that co-culture with enteroids resulted in a loss of RALDH activity in 'gut-like' DCs. Continued provision of GM-CSF and RA during co-culture was required to oppose putative negative signals from the enteroid epithelium. Our data contribute to a growing understanding of how intestinal cDCs assess environmental conditions to ensure appropriate activation of the immune response.

20.
FASEB J ; 25(6): 1856-64, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21350119

ABSTRACT

In intact mucosal tissues, epithelial cells are anatomically positioned in proximity to a number of subepithelial cell types, including endothelia. A number of recent studies have suggested that imbalances between energy supply and demand can result in "inflammatory hypoxia." Given these associations, we hypothesized that endothelial-derived, hypoxia-inducible mediators might influence epithelial function. Guided by cDNA microarray analysis of human microvascular endothelial cells (HMEC-1 line) subjected to hypoxia (pO(2) 20 torr, 8 h), we identified adrenomedullin (ADM) as a prominent hypoxia-inducible factor (HIF) that acts on epithelial cells through cell surface receptors. We assessed the functional ability for exogenous ADM to signal in human intestinal Caco2 cells in vitro by demonstrating a dose-dependent induction of Erk1/2phosphorylation. Further analysis revealed that ADM deneddylates cullin-2 (Cul2), whose action has been demonstrated to control the activity of HIF. Caco2 cells stably expressing a hypoxic response element (HRE)-driven luciferase promoter confirmed that ADM activates the HIF signaling pathway. Extensions of these studies revealed an increase in canonical HIF-1-dependent genes following stimulation with ADM. To define physiological relevance, we investigated the effect of ADM in a DSS model of murine colitis. Administration of ADM resulted in reduced inflammatory indices and less severe histological inflammation compared to vehicle controls. Analysis of tissue and serum cytokines showed a marked and significant inhibition of colitis-associated TNF-α, IL-1ß, and KC. Analysis of circulating ADM demonstrated an increase in serum ADM in murine models of colitis. Taken together, these results identify ADM as an endogenously generated vascular mediator that functions as a mucosal protective factor through fine tuning of HIF activity.


Subject(s)
Adrenomedullin/physiology , Endothelial Cells/metabolism , Hypoxia-Inducible Factor 1/metabolism , Inflammation/metabolism , Adrenomedullin/metabolism , Adrenomedullin/pharmacology , Animals , Caco-2 Cells , Cell Line , Colitis/chemically induced , Colitis/drug therapy , Dextran Sulfate/toxicity , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Regulation/physiology , Humans , Hypoxia-Inducible Factor 1/genetics , Intestinal Mucosa/cytology , Mice , Mice, Inbred C57BL , Oxygen Consumption , Receptors, Adrenomedullin/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL