Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 34(3): 3732-3742, 2020 03.
Article in English | MEDLINE | ID: mdl-31944416

ABSTRACT

The HIF hydroxylase enzymes (PHD1-3 and FIH) are cellular oxygen-sensors which confer hypoxic-sensitivity upon the hypoxia-inducible factors HIF-1α and HIF-2α. Microenvironmental hypoxia has a strong influence on the epithelial and immune cell function through HIF-dependent gene expression and consequently impacts upon the course of disease progression in ulcerative colitis (UC), with HIF-1α being protective while HIF-2α promotes disease. However, little is known about how inflammation regulates hypoxia-responsive pathways in UC patients. Here we demonstrate that hypoxia is a prominent microenvironmental feature of the mucosa in UC patients with active inflammatory disease. Furthermore, we found that inflammation drives transcriptional programming of the HIF pathway including downregulation of PHD1 thereby increasing the tissue responsiveness to hypoxia and skewing this response toward protective HIF-1 over detrimental HIF-2 activation. We identified CEBPα as a transcriptional regulator of PHD1 mRNA expression which is downregulated in both inflamed tissue derived from patients and in cultured intestinal epithelial cells treated with inflammatory cytokines. In summary, we propose that PHD1 downregulation skews the hypoxic response toward enhanced protective HIF-1α stabilization in the inflamed mucosa of UC patients.


Subject(s)
Colitis, Ulcerative/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Inflammation/metabolism , Blotting, Western , Caco-2 Cells , Chromatin Immunoprecipitation , Colitis, Ulcerative/genetics , Computational Biology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Immunohistochemistry , Inflammation/genetics , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
2.
Inflamm Bowel Dis ; 26(2): 192-205, 2020 01 06.
Article in English | MEDLINE | ID: mdl-31618435

ABSTRACT

BACKGROUND: Pouchitis is the most common long-term complication after restorative proctocolectomy with ileal pouch-anal anastomosis (IPAA) for ulcerative colitis (UC) or familial adenomatous polyposis (FAP), which can eventually progress to pouch failure, necessitating permanent stoma construction. Hypoxia-inducible transcription factor prolyl hydroxylase-containing enzymes (PHD1, PHD2, and PHD3) are molecular oxygen sensors that control adaptive gene expression through hypoxia-inducible factor (HIF). Emerging evidence supports PHDs as being therapeutic targets in intestinal inflammation. However, pharmacological inhibition of PHDs has not been validated as a treatment strategy in pouchitis. METHODS: PHD1-3 mRNA and protein expression were analyzed in mucosal pouch and prepouch ileal patient biopsies. After establishment of a preclinical IPAA model in rats, the impact of the pan-PHD small-molecule inhibitor dimethyloxalylglycine (DMOG) on dextran sulfate sodium (DSS)-induced pouchitis was studied. Clinical and molecular parameters were investigated. RESULTS: PHD1, but not PHD2 or PHD3, was overexpressed in pouchitis in biopsies of patients with IPAA for UC but not FAP. In addition, PHD1 expression correlated with disease activity. DMOG treatment profoundly mitigated DSS-induced pouchitis in a rodent IPAA model. Mechanistically, DMOG restored intestinal epithelial barrier function by induction of tight junction proteins zona occludens-1 and claudin-1 and alleviation of intestinal epithelial cell apoptosis, thus attenuating pouch inflammation. CONCLUSIONS: Together, these results establish a strong therapeutic rationale for targeting PHD1 with small-molecule inhibitors in pouchitis after IPAA for UC.


Subject(s)
Pouchitis/prevention & control , Prolyl Hydroxylases/chemistry , Prolyl-Hydroxylase Inhibitors/therapeutic use , Animals , Humans , Pouchitis/enzymology , Pouchitis/pathology
3.
Am J Physiol Gastrointest Liver Physiol ; 317(2): G90-G97, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31070931

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by epithelial barrier dysfunction with resultant inflammation as the mucosal immune system becomes exposed to luminal antigens. The hydroxylase inhibitor dimethyloxalylglycine (DMOG) reduces symptoms in experimental colitis through the upregulation of genes promoting barrier function and inhibition of epithelial cell apoptosis. The immunosuppressive drug cyclosporine reduces inflammation associated with IBD via suppression of immune cell activation. Given the distinct barrier protective effect of DMOG and the anti-inflammatory properties of cyclosporine, we hypothesized that combining these drugs may provide an enhanced protective effect by targeting both barrier dysfunction and inflammation simultaneously. We used the dextran sulfate sodium model of colitis in C57BL/6 mice to determine the combinatorial efficacy of cyclosporine and DMOG. While cyclosporine and DMOG ameliorated disease progression, in combination they had an additive protective effect that surpassed the level of protection afforded by either drug alone. The ability of DMOG to augment the anti-inflammatory effects of cyclosporine was largely due to preservation of barrier function and at least in part due to zonula occludens-1 regulation. We propose that combining the barrier protective effects of a hydroxylase inhibitor with the anti-inflammatory effects of cyclosporine provides added therapeutic benefit in colitis.NEW & NOTEWORTHY Inflammatory bowel disease is the result of decreased intestinal epithelial barrier function leading to exposure of the mucosal immune system to luminal antigens causing inflammation, which in turn further decreases epithelial barrier function. We demonstrate for the first time that strengthening the epithelial barrier with a hydroxylase inhibitor in combination with the administration of the immunosuppressive cyclosporine provides additive therapeutic advantage in a murine model of colitis.


Subject(s)
Amino Acids, Dicarboxylic/pharmacology , Colitis , Cyclosporine/pharmacology , Hypoxia/immunology , Intestinal Mucosa , Animals , Apoptosis/drug effects , Colitis/immunology , Colitis/physiopathology , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination/methods , Hypoxia-Inducible Factor 1/metabolism , Immunosuppressive Agents/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Protective Agents/pharmacology , Up-Regulation
4.
J Immunol ; 202(5): 1521-1530, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30700584

ABSTRACT

Hypoxia is a common and prominent feature of the microenvironment at sites of bacteria-associated inflammation in inflammatory bowel disease. The prolyl-hydroxylases (PHD1/2/3) and the asparaginyl-hydroxylase factor-inhibiting HIF are oxygen-sensing enzymes that regulate adaptive responses to hypoxia through controlling the activity of HIF and NF-κB-dependent transcriptional pathways. Previous studies have demonstrated that the pan-hydroxylase inhibitor dimethyloxalylglycine (DMOG) is effective in the alleviation of inflammation in preclinical models of inflammatory bowel disease, at least in part, through suppression of IL-1ß-induced NF-κB activity. TLR-dependent signaling in immune cells, such as monocytes, which is important in bacteria-driven inflammation, shares a signaling pathway with IL-1ß. In studies into the effect of pharmacologic hydroxylase inhibition on TLR-induced inflammation in monocytes, we found that DMOG selectively triggers cell death in cultured THP-1 cells and primary human monocytes at concentrations well tolerated in other cell types. DMOG-induced apoptosis was independent of increased caspase-3/7 activity but was accompanied by reduced expression of the inhibitor of apoptosis protein 1 (cIAP1). Based on these data, we hypothesize that pharmacologic inhibition of the HIF-hydroxylases selectively targets monocytes for cell death and that this may contribute to the anti-inflammatory activity of HIF-hydroxylase inhibitors.


Subject(s)
Amino Acids, Dicarboxylic/pharmacology , Inflammation/drug therapy , Mixed Function Oxygenases/antagonists & inhibitors , Monocytes/drug effects , Prolyl-Hydroxylase Inhibitors/pharmacology , Cell Death/drug effects , Cell Death/immunology , Cells, Cultured , HEK293 Cells , Humans , Inflammation/immunology , Inflammation/metabolism , Mixed Function Oxygenases/immunology , Mixed Function Oxygenases/metabolism , Monocytes/immunology , Monocytes/metabolism
5.
Allergy ; 74(4): 753-766, 2019 04.
Article in English | MEDLINE | ID: mdl-30394557

ABSTRACT

BACKGROUND: When an immune cell migrates from the bloodstream to a site of chronic inflammation, it experiences a profound decrease in microenvironmental oxygen levels leading to a state of cellular hypoxia. The hypoxia-inducible factor-1α (HIF-1α) promotes an adaptive transcriptional response to hypoxia and as such is a major regulator of immune cell survival and function. HIF hydroxylases are the family of oxygen-sensing enzymes primarily responsible for conferring oxygen dependence upon the HIF pathway. METHODS: Using a mouse model of allergic contact dermatitis (ACD), we tested the effects of treatment with the pharmacologic hydroxylase inhibitor DMOG, which mimics hypoxia, on disease development. RESULTS: Re-exposure of sensitized mice to 2,4-dinitrofluorobenzene (DNFB) elicited inflammation, edema, chemokine synthesis (including CXCL1 and CCL5) and the recruitment of neutrophils and eosinophils. Intraperitoneal or topical application of the pharmacologic hydroxylase inhibitors dymethyloxalylglycine (DMOG) or JNJ1935 attenuated this inflammatory response. Reduced inflammation was associated with diminished recruitment of neutrophils and eosinophils but not lymphocytes. Finally, hydroxylase inhibition reduced cytokine-induced chemokine production in cultured primary keratinocytes through attenuation of the JNK pathway. CONCLUSION: These data demonstrate that hydroxylase inhibition attenuates the recruitment of neutrophils to inflamed skin through reduction of chemokine production and increased neutrophilic apoptosis. Thus, pharmacologic inhibition of HIF hydroxylases may be an effective new therapeutic approach in allergic skin inflammation.


Subject(s)
Amino Acids, Dicarboxylic/therapeutic use , Dermatitis, Allergic Contact/prevention & control , Mixed Function Oxygenases/antagonists & inhibitors , Amino Acids, Dicarboxylic/pharmacology , Animals , Cell Movement/drug effects , Cytokines/metabolism , Eosinophils/cytology , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit , Inflammation/drug therapy , Mice , Neutrophils/cytology
6.
Semin Immunol ; 28(5): 469-477, 2016 10.
Article in English | MEDLINE | ID: mdl-27717536

ABSTRACT

Crosstalk between metabolic and immune pathways has recently become appreciated to be key to the regulation of host defence. The hypoxia-inducible factor (HIF) is a transcription factor which was initially described as a ubiquitous master regulator of the transcriptional response to hypoxia. In this role, HIF regulates genes promoting adaptation to hypoxia including a number which influence the cellular metabolic strategy of a cell. It has more recently been appreciated that the regulation of HIF is not restricted to oxygen-dependent pathways, and is now known to be mediated by a number of additional metabolic and immune cues including metabolites and cytokines respectively. Furthermore, our understanding of the functional role of HIF has expanded to it now being appreciated as a major regulator of host immunity. This places HIF in an ideal position to act as a regulatory hub which links metabolic activity with immunity. In this review we synthesise recent data which identifies HIF as both a target and effector for metabolic and immune processes. Developing our understanding of the role of HIF in this context will uncover new therapeutic targets for inflammatory and infectious disease.


Subject(s)
Energy Metabolism , Hypoxia-Inducible Factor 1/metabolism , Immunity , Animals , Energy Metabolism/drug effects , Gene Expression Regulation/drug effects , Humans , Hypoxia/genetics , Hypoxia/immunology , Hypoxia/metabolism , Immunomodulation , Inflammation/drug therapy , Inflammation/etiology , Inflammation/metabolism , Mixed Function Oxygenases/antagonists & inhibitors , Molecular Targeted Therapy , Signal Transduction/drug effects
7.
PLoS Biol ; 14(1): e1002347, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26752685

ABSTRACT

The asparagine hydroxylase, factor inhibiting HIF (FIH), confers oxygen-dependence upon the hypoxia-inducible factor (HIF), a master regulator of the cellular adaptive response to hypoxia. Studies investigating whether asparagine hydroxylation is a general regulatory oxygen-dependent modification have identified multiple non-HIF targets for FIH. However, the functional consequences of this outside of the HIF pathway remain unclear. Here, we demonstrate that the deubiquitinase ovarian tumor domain containing ubiquitin aldehyde binding protein 1 (OTUB1) is a substrate for hydroxylation by FIH on N22. Mutation of N22 leads to a profound change in the interaction of OTUB1 with proteins important in cellular metabolism. Furthermore, in cultured cells, overexpression of N22A mutant OTUB1 impairs cellular metabolic processes when compared to wild type. Based on these data, we hypothesize that OTUB1 is a target for functional hydroxylation by FIH. Additionally, we propose that our results provide new insight into the regulation of cellular energy metabolism during hypoxic stress and the potential for targeting hydroxylases for therapeutic benefit.


Subject(s)
Cysteine Endopeptidases/metabolism , Mixed Function Oxygenases/metabolism , Repressor Proteins/metabolism , AMP-Activated Protein Kinases/metabolism , Cysteine Endopeptidases/genetics , Deubiquitinating Enzymes , Energy Metabolism , HEK293 Cells , Humans , Hydroxylation , Mutagenesis, Site-Directed , Protein Stability
SELECTION OF CITATIONS
SEARCH DETAIL
...