1
|
Peroxiredoxins and Hypoxia-Inducible Factor-1α in Duodenal Tissue: Emerging Factors in the Pathophysiology of Pediatric Celiac Disease Patients. Curr Issues Mol Biol 2023; 45:1779-1793. [PMID: 36826059 PMCID: PMC9954839 DOI: 10.3390/cimb45020114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023] Open
Abstract
Celiac disease (CD) is an autoimmune enteropathy. Peroxiredoxins (PRDXs) are powerful antioxidant enzymes having an important role in significant cellular pathways including cell survival, apoptosis, and inflammation. This study aimed at investigating the expression levels of all PRDX isoforms (1-6) and their possible relationships with a transcription factor, HIF-1α, in the small intestinal tissue samples of pediatric CD patients. The study groups consisted of first-diagnosed CD patients (n = 7) and non-CD patients with functional gastrointestinal tract disorders as the controls (n = 7). The PRDXs and HIF-1α expression levels were determined by using real-time PCR and Western blotting in duodenal biopsy samples. It was observed that the mRNA and protein expression levels of PRDX 5 were significantly higher in the CD patients, whereas the PRDX 1, -2, and -4 expressions were decreased in each case compared to the control group. No significant differences were detected in the PRDX 3 and PRDX 6 expressions. The expression of HIF-1α was also significantly elevated in CD patients. These findings indicate, for the first time, that PRDXs, particularly PRDX 5, may play a significant role in the pathogenesis of CD. Furthermore, our results suggest that HIF-1α may upregulate PRDX-5 transcription in the duodenal tissue of CD.
Collapse
|
2
|
Maddah R, Panji A, Khoei MA, Bazireh H, Abedinlou H, Davodabadi F, Shariati P. Deciphering potential biomarkers for celiac disease by using an integrated bioinformatics approach. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.101040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022] Open
|
3
|
Drozdzik M, Czekawy I, Oswald S, Drozdzik A. Intestinal drug transporters in pathological states: an overview. Pharmacol Rep 2020; 72:1173-1194. [PMID: 32715435 PMCID: PMC7550293 DOI: 10.1007/s43440-020-00139-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Affiliation(s)
- Marek Drozdzik
- Department of Pharmacology, Pomeranian Medical University, Powstancow Wlkp 72, 70-111, Szczecin, Poland.
| | - Izabela Czekawy
- Department of Pharmacology, Pomeranian Medical University, Powstancow Wlkp 72, 70-111, Szczecin, Poland
| | - Stefan Oswald
- Department of Pharmacology, Medicine University Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489, Greifswald, Germany.,Institute of Pharmacology and Toxicology, Rostock University Medical Center, 18051, Rostock, Germany
| | - Agnieszka Drozdzik
- Department of Integrated Dentistry, Pomeranian Medical University, Powstancow Wlkp 72, 70-111, Szczecin, Poland
| |
Collapse
|
4
|
Kumar T, Pandey R, Chauhan NS. Hypoxia Inducible Factor-1α: The Curator of Gut Homeostasis. Front Cell Infect Microbiol 2020; 10:227. [PMID: 32500042 PMCID: PMC7242652 DOI: 10.3389/fcimb.2020.00227] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/23/2020] [Indexed: 12/11/2022] Open
Abstract
The human gut microbiome is a stratified and resilient ecosystem co-inhabited by a diverse and dynamic pool of microorganisms. Microbial selection, establishment, and colonization are modulated through a complex molecular network of host-microbial interactions. These molecular bioprocesses ensure the taxonomic composition of the mature human gut microbiome. The human gut microbiome plays a vital role in host health; otherwise, any microbial dysbiosis could predispose to the onset of physiological and metabolic disorder/s. Focussed research are being carried out to identify key molecular agents defining gut homeostasis. These molecules hold the potential to develop effective therapeutic solutions for microbial dysbiosis-associated human disorders. Of these, Hypoxia-inducible factor-1α (HIF-1α) is a central player in host-microbial crosstalk to maintain gut homeostasis. Human gut microbial metabolites regulate its cellular stability, which in turn regulates various cellular processes required for the stable gut microbiome. In the present review, an effort has been made to summarize the key role of HIF-1α to maintain gut homeostasis.
Collapse
Affiliation(s)
- Tarun Kumar
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, India
| | - Rajesh Pandey
- Genomics and Molecular Medicine, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
| | - Nar Singh Chauhan
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, India
| |
Collapse
|
5
|
Maurano F, Ogita T, Luongo D, Rotondi Aufiero V, Bergamo P, Mazzarella G, Tanabe S, Rossi M. Innate immunity is a late event in the onset of gliadin-specific enteropathy in the HLA-DQ8 mice. Immunobiology 2020; 225:151903. [PMID: 31928782 DOI: 10.1016/j.imbio.2020.151903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/05/2020] [Indexed: 12/30/2022]
Abstract
Celiac disease (CD) is a food enteropathy that occurs in genetically susceptible individuals following the ingestion of gluten. Both gluten cytotoxicity and immunity activation play a role in CD pathogenesis; however, the chronological assessment of the different pathogenic mechanisms remains elusive. The models developed so far have only partially addressed this issue. Herein, Ab°DQ8 transgenic mice were administered wheat gliadin and indomethacin for 10 days to induce enteropathy. Gliadin-induced alteration of the small intestinal architecture was associated with increased expression of tissue transglutaminase in the lamina propria and a marked hypoxic environment. Enteropathic mice showed activation of innate immunity, featuring an increase of pro-inflammatory IFN-γ and IL-15 mRNAs, as well as CD11c+CD103+, CD11b+CD11c+, and CD11b+CD103+ dendritic cell subsets. However, the temporal assessment of examined parameters indicated that the induction of innate immunity during the generation of the mucosal lesion, occurred belatedly, highlighting a major role of gliadin intrinsic cytotoxicity in the pathogenic mechanism of this model. These results have important implications for the use of this model to test the impact of biotechnological interventions to reduce the cytotoxicity of gliadin.
Collapse
Affiliation(s)
- Francesco Maurano
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Tasuku Ogita
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minamiminowa, Kamiina, Nagano 399-4598, Japan
| | - Diomira Luongo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | | | - Paolo Bergamo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | | | - Soichi Tanabe
- Hiroshima University, Graduate School of Biosphere Science, Hiroshima, Japan
| | - Mauro Rossi
- Institute of Food Sciences, National Research Council, Avellino, Italy.
| |
Collapse
|
6
|
Caminero A, McCarville JL, Zevallos VF, Pigrau M, Yu XB, Jury J, Galipeau HJ, Clarizio AV, Casqueiro J, Murray JA, Collins SM, Alaedini A, Bercik P, Schuppan D, Verdu EF. Lactobacilli Degrade Wheat Amylase Trypsin Inhibitors to Reduce Intestinal Dysfunction Induced by Immunogenic Wheat Proteins. Gastroenterology 2019; 156:2266-2280. [PMID: 30802444 DOI: 10.1053/j.gastro.2019.02.028] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 02/08/2019] [Accepted: 02/19/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Wheat-related disorders, a spectrum of conditions induced by the ingestion of gluten-containing cereals, have been increasing in prevalence. Patients with celiac disease have gluten-specific immune responses, but the contribution of non-gluten proteins to symptoms in patients with celiac disease or other wheat-related disorders is controversial. METHODS C57BL/6 (control), Myd88-/-, Ticam1-/-, and Il15-/- mice were placed on diets that lacked wheat or gluten, with or without wheat amylase trypsin inhibitors (ATIs), for 1 week. Small intestine tissues were collected and intestinal intraepithelial lymphocytes (IELs) were measured; we also investigated gut permeability and intestinal transit. Control mice fed ATIs for 1 week were gavaged daily with Lactobacillus strains that had high or low ATI-degrading capacity. Nonobese diabetic/DQ8 mice were sensitized to gluten and fed an ATI diet, a gluten-containing diet or a diet with ATIs and gluten for 2 weeks. Mice were also treated with Lactobacillus strains that had high or low ATI-degrading capacity. Intestinal tissues were collected and IELs, gene expression, gut permeability and intestinal microbiota profiles were measured. RESULTS In intestinal tissues from control mice, ATIs induced an innate immune response by activation of Toll-like receptor 4 signaling to MD2 and CD14, and caused barrier dysfunction in the absence of mucosal damage. Administration of ATIs to gluten-sensitized mice expressing HLA-DQ8 increased intestinal inflammation in response to gluten in the diet. We found ATIs to be degraded by Lactobacillus, which reduced the inflammatory effects of ATIs. CONCLUSIONS ATIs mediate wheat-induced intestinal dysfunction in wild-type mice and exacerbate inflammation to gluten in susceptible mice. Microbiome-modulating strategies, such as administration of bacteria with ATI-degrading capacity, may be effective in patients with wheat-sensitive disorders.
Collapse
Affiliation(s)
- Alberto Caminero
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Justin L McCarville
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Victor F Zevallos
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Marc Pigrau
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Xuechen B Yu
- Department of Medicine, Columbia University, New York, New York; Institute of Human Nutrition, Columbia University, New York, New York
| | - Jennifer Jury
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Heather J Galipeau
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Alexandra V Clarizio
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | | | - Joseph A Murray
- Division of Gastroenterology and Hepatology, Department of Immunology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Stephen M Collins
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Armin Alaedini
- Department of Medicine, Columbia University, New York, New York; Institute of Human Nutrition, Columbia University, New York, New York
| | - Premysl Bercik
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Elena F Verdu
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
7
|
Effinger A, O'Driscoll CM, McAllister M, Fotaki N. Impact of gastrointestinal disease states on oral drug absorption – implications for formulation design – a PEARRL review. J Pharm Pharmacol 2018; 71:674-698. [PMID: 29766501 DOI: 10.1111/jphp.12928] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 04/16/2018] [Indexed: 12/18/2022]
Abstract
Abstract
Objectives
Drug product performance in patients with gastrointestinal (GI) diseases can be altered compared to healthy subjects due to pathophysiological changes. In this review, relevant differences in patients with inflammatory bowel diseases, coeliac disease, irritable bowel syndrome and short bowel syndrome are discussed and possible in vitro and in silico tools to predict drug product performance in this patient population are assessed.
Key findings
Drug product performance was altered in patients with GI diseases compared to healthy subjects, as assessed in a limited number of studies for some drugs. Underlying causes can be observed pathophysiological alterations such as the differences in GI transit time, the composition of the GI fluids and GI permeability. Additionally, alterations in the abundance of metabolising enzymes and transporter systems were observed. The effect of the GI diseases on each parameter is not always evident as it may depend on the location and the state of the disease. The impact of the pathophysiological change on drug bioavailability depends on the physicochemical characteristics of the drug, the pharmaceutical formulation and drug metabolism. In vitro and in silico methods to predict drug product performance in patients with GI diseases are currently limited but could be a useful tool to improve drug therapy.
Summary
Development of suitable in vitro dissolution and in silico models for patients with GI diseases can improve their drug therapy. The likeliness of the models to provide accurate predictions depends on the knowledge of pathophysiological alterations, and thus, further assessment of physiological differences is essential.
Collapse
Affiliation(s)
- Angela Effinger
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | | | | | - Nikoletta Fotaki
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| |
Collapse
|
8
|
Rodieux F, Gotta V, Pfister M, van den Anker JN. Causes and Consequences of Variability in Drug Transporter Activity in Pediatric Drug Therapy. J Clin Pharmacol 2017; 56 Suppl 7:S173-92. [PMID: 27385174 DOI: 10.1002/jcph.721] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/26/2016] [Accepted: 02/11/2016] [Indexed: 01/06/2023]
Abstract
Drug transporters play a key role in mediating the uptake of endo- and exogenous substances into cells as well as their efflux. Therefore, variability in drug transporter activity can influence pharmaco- and toxicokinetics and be a determinant of drug safety and efficacy. In children, particularly in neonates and young infants, the contribution of tissue-specific drug transporters to drug absorption, distribution, and excretion may differ from that in adults. In this review 5 major factors and their interdependence that may influence drug transporter activity in children are discussed: developmental differences, genetic polymorphisms, pediatric comorbidities, interacting comedication, and environmental factors. Even if data are sparse, altered drug transporter activity due to those factors have been associated with clinically relevant differences in drug disposition, efficacy, and safety in pediatric patients. Single nucleotide polymorphisms in drug transporter-encoding genes were the most studied source of drug transporter variability in children. However, in the age group where drug transporter activity has been reported to differ from that in adults, namely neonates and young infants, hardly any studies have been performed. Longitudinal studies in this young population are required to investigate the age- and disease-dependent genotype-phenotype relationships and relevance of drug transporter drug-drug interactions. Physiologically based pharmacokinetic modeling approaches can integrate drug- and patient-specific parameters, including drug transporter ontogeny, and may further improve in silico predictions of pediatric-specific pharmacokinetics.
Collapse
Affiliation(s)
- Frédérique Rodieux
- Pediatric Pharmacology, University of Basel Children's Hospital (UKBB), Basel, Switzerland
| | - Verena Gotta
- Pediatric Pharmacology, University of Basel Children's Hospital (UKBB), Basel, Switzerland
| | - Marc Pfister
- Pediatric Pharmacology, University of Basel Children's Hospital (UKBB), Basel, Switzerland.,Quantitative Solutions/Certara, Menlo Park, CA, USA
| | - Johannes N van den Anker
- Pediatric Pharmacology, University of Basel Children's Hospital (UKBB), Basel, Switzerland.,Division of Pediatric Clinical Pharmacology, Children's National Health System, Washington, DC, USA.,Intensive Care and Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
9
|
Sziksz E, Molnár K, Lippai R, Pap D, Onody A, Veres-Székely A, Vörös P, Szabó D, Győrffy H, Veres G, Tulassay T, Vannay A, Arató A. Peroxisome proliferator-activated receptor-γ and thymic stromal lymphopoietin are involved in the pathophysiology of childhood coeliac disease. Virchows Arch 2014; 465:385-93. [PMID: 25187315 DOI: 10.1007/s00428-014-1650-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 08/01/2014] [Accepted: 08/26/2014] [Indexed: 12/22/2022]
Abstract
Celiac disease (CD) is a chronic autoimmune enteropathy caused by exposure to dietary gluten in genetically predisposed individuals. The transcription factor peroxisome proliferator-activated receptor gamma (PPARγ) was shown to exert protective effects in several immune-mediated disorders. Activation of PPARγ suppressed the expression of thymic stromal lymphopoietin (TSLP), an inducer of proinflammatory cytokines. Since the role of TSLP in gluten-sensitive enteropathy is completely unknown, we investigated the involvement of TSLP and its regulator PPARγ in childhood CD. We collected duodenal biopsy specimens from 19 children with newly diagnosed CD, 6 children with treated CD (gluten-free diet, GFD), and 10 controls. Expression of mRNA and protein levels of PPARγ, TSLP, and TSLP receptor were determined by real-time RT-PCR and Western blot, respectively. Duodenal localization of PPARγ and TSLP was studied by immunohistochemistry. In duodenal mucosa of children with CD, the amount of PPARγ was significantly lower and simultaneously that of TSLP significantly higher compared to controls (p < 0.05). In GFD-treated patients, the levels of PPARγ mRNA and protein were significantly higher while that of TSLP markedly lower compared to newly diagnosed CD (p < 0.05). Immunohistochemistry revealed PPARγ and TSLP expression in lamina propria immune cells and in enterocytes. Low expression of PPARγ and high expression of TSLP in the duodenal mucosa of children with newly diagnosed CD suggest that they are involved in the pathophysiology of CD. We hypothesize that PPARγ may be an inhibitory regulator of TSLP-stimulated inflammatory processes in CD.
Collapse
Affiliation(s)
- Erna Sziksz
- MTA-SE, Pediatrics and Nephrology Research Group, Budapest, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Wang WS, Liang HY, Cai YJ, Yang H. DMOG ameliorates IFN-γ-induced intestinal barrier dysfunction by suppressing PHD2-dependent HIF-1α degradation. J Interferon Cytokine Res 2013; 34:60-9. [PMID: 24010824 DOI: 10.1089/jir.2013.0040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Hypoxia-inducible factor 1α (HIF-1α) has been well established as a protective factor for intestinal barrier function in intestinal epithelial cells. Recently, a study found that increased HIF-1α-induced intestinal barrier dysfunction. We proposed that lymphocyte-derived interferon-gamma (IFN-γ) might be responsible for the intestinal barrier dysfunction caused by increased HIF-1α. HT-29 cell monolayers were grown in the presence or absence of IFN-γ under hypoxia. Then, the transepithelial electrical resistance was measured, and HIF-1α-modulated intestinal barrier protective factors were quantified by polymerase chain reaction (PCR). PCR, western blotting, and chromatin immunoprecipitation of HIF-1α were performed. Dimethyloxalyglycine (DMOG), an inhibitor of prolyl-hydroxylases (PHDs) that stabilizes HIF-1α during normoxia, and RNA interference of PHDs were also used to identify the signal pathway between IFN-γ and HIF-1α. We demonstrated that IFN-γ caused barrier dysfunction in hypoxic HT-29 cell monolayers via suppressing HIF-1α and HIF-1α-modulated intestinal barrier protective factors. We found that IFN-γ decreased HIF-1α protein expression instead of affecting HIF-1α transcription or transcriptional activity. Study also showed that DMOG reversed the IFN-γ-induced decrease in HIF-1α protein expression. Further, we found that PHD2 is the major regulator of IFN-γ-induced HIF-1α degradation by PHD inhibition and RNA interference. We conclude that IFN-γ caused barrier dysfunction by promoting PHD-, especially PHD2-, dependent HIF-1α degradation, and DMOG or PHD2 inhibition reversed this HIF-1α suppression and ameliorated barrier dysfunction. Combined with other studies demonstrating HIF-1α activation in lymphocytes promotes IFN-γ secretion, these findings suggest a mechanism by which increased HIF-1α-induced intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Wen-Sheng Wang
- 1 Department of General Surgery, Xinqiao Hospital, Third Military Medical University , Chongqing, China
| | | | | | | |
Collapse
|
11
|
Vörös P, Sziksz E, Himer L, Onody A, Pap D, Frivolt K, Szebeni B, Lippai R, Győrffy H, Fekete A, Brandt F, Molnár K, Veres G, Arató A, Tulassay T, Vannay A. Expression of PARK7 is increased in celiac disease. Virchows Arch 2013; 463:401-8. [PMID: 23832581 DOI: 10.1007/s00428-013-1443-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 05/14/2013] [Accepted: 06/19/2013] [Indexed: 02/07/2023]
Abstract
Recently, it has been suggested that the gene called Parkinson's disease 7 (PARK7) might be an upstream activator of hypoxia-inducible factor (HIF)-1α, which plays a major role in sustaining intestinal barrier integrity. Furthermore, PARK7 has been proposed to participate in the Toll-like receptor (TLR)-dependent regulation of the innate immune system. Our aim was to investigate the involvement of PARK7 in the pathogenesis of coeliac disease (CD). Duodenal biopsy specimens were collected from 19 children with untreated CD, five children with treated CD (maintained on gluten-free diet), and ten children with histologically normal duodenal biopsies. PARK7 mRNA expression and protein level were determined by real-time polymerase chain reaction (PCR) and Western blot, respectively. Localization of PARK7 was visualized by immunofluorescence staining. Protein level of PARK7 increased in the duodenal mucosa of children with untreated CD compared to children with treated CD or to control biopsies (p <0.03). We detected intensive PARK7 staining in the epithelial cells and lamina propria of the duodenal mucosa of children with untreated CD compared with that in control biopsies. Our finding that mucosal expression of PARK7 is increased suggests that PARK7 is involved in the pathogenesis of gastrointestinal diseases, notably CD. Our results suggest that PARK7 may alter processes mediated by HIF-1α and TLR4, which supports a role for PARK7 in the maintenance of epithelial barrier integrity, immune homeostasis, or apoptosis.
Collapse
Affiliation(s)
- Péter Vörös
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Scholz CC, Taylor CT. Targeting the HIF pathway in inflammation and immunity. Curr Opin Pharmacol 2013; 13:646-53. [PMID: 23660374 DOI: 10.1016/j.coph.2013.04.009] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 04/12/2013] [Accepted: 04/15/2013] [Indexed: 01/03/2023]
Abstract
Oxygen deprivation (hypoxia) is a frequently encountered condition in both health and disease. Metazoans have evolved an elegant and direct cellular mechanism by which to sense local oxygen levels and mount an adaptive transcriptional response to hypoxia which is mediated by a transcription factor termed the hypoxia-inducible factor (HIF). In normoxia, HIF is repressed primarily through the action of a family of hydroxylases, which target HIFα subunits for degradation in an oxygen-dependent manner. In hypoxia, HIF is rapidly stabilized in cells thus allowing it to regulate the expression of hundreds of genes which promote an adaptive response including genes expressing regulators of angiogenesis, metabolism, growth and survival. Initial studies into the HIF pathway focused mainly on its role in supporting tumor adaptation through enhancing processes such as angiogenesis, glycolytic metabolism and cell survival. More recently however, it has become clear that the HIF pathway also plays a key role in the regulation of immunity and inflammation. In fact, conditional knockout of the HIF-1α subunit has identified key immune roles in T-cells, dendritic cells, macrophages, neutrophils and epithelial cells. In this review, we will consider the role for HIF in the regulation of the immune response and its possible contribution to inflammation. Furthermore, we will consider potential therapeutic strategies, which target the HIF pathway in chronic inflammatory and infectious disease.
Collapse
Affiliation(s)
- Carsten C Scholz
- Systems Biology Ireland, School of Medicine and Medical Science & The Conway Institute, University College Dublin, Ireland
| | | |
Collapse
|
13
|
Abstract
Inflammatory bowel disease (IBD) is a common and debilitating clinical disorder comprising ulcerative colitis and Crohn's disease. IBD occurs when inappropriate immunological activity in the intestinal mucosa results in epithelial barrier dysfunction leading to exposure of the mucosal immune system to luminal antigenic material. This in turn results in the cycles of inflammation and further barrier dysfunction which underlie disease progression. Although significant therapeutic advances have been made over the last decade, current immunosuppressive and anti-inflammatory treatments for IBD have significant limitations due to lack of treatment response in some patients and adverse effects, including increased risk of infection and malignancy. Recent studies using experimental models of IBD have identified that intracellular hydroxylases, a group of enzymes responsible for oxygen sensing and activation of adaptive transcriptional responses to hypoxia may represent a new class of therapeutic targets in IBD. Hydroxylase inhibitors are effective in ameliorating symptoms of colitis at least in part through the promotion of intestinal epithelial barrier function. The mechanism of this protection is due to activation of hypoxia-sensitive transcription factors, including the hypoxia-inducible factor (HIF) and nuclear factor kappa-B (NF-κB), which activate specific epithelial barrier-protective transcriptional programs. In this review, the mechanism(s) of action and the therapeutic potential of small molecule hydroxylase inhibitors for the treatment of IBD will be discussed.
Collapse
|
14
|
Mimouna S, Gonçalvès D, Barnich N, Darfeuille-Michaud A, Hofman P, Vouret-Craviari V. Crohn disease-associated Escherichia coli promote gastrointestinal inflammatory disorders by activation of HIF-dependent responses. Gut Microbes 2013; 2:335-46. [PMID: 22157238 DOI: 10.4161/gmic.18771] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Crohn disease (CD) ileal lesions are colonized by adherent-invasive E. coli (AIEC) that locally induce inflammation. Hypoxia inducible factor (HIF)-1alpha protein is expressed in acute and chronically inflamed site; however the molecular basis of this expression is not fully understood. The aim of the study was to access whether AIEC induce HIF-1α expression and to study the consequence of HIF-1α expression on the onset of Crohn disease pathogenesis. We show that HIF-1α is maximally expressed in inflamed ileal epithelium of CD-patients. CEACAM6, a protein that acts as a receptor of AIEC, is expressed in this particular condition. Using CEABAC 10 transgenic mice that express CEACAM6, we show that AIEC bacteria, but not non-pathogenic E. coli K12, induce the production of HIF-1alpha protein and the activation of VEGF/VEGFR signaling. Downstream analyses on human intestinal epithelial cells silenced for hif- 1α, highlight the crucial role of this protein in production of pro-angiogenic factors. This study highlights the crucial role of AIEC bacteria as promoter of inflammatory disorders of the gastrointestinal tract and provides clear evidence that HIF-1α protein plays a major role in mediating this effect.
Collapse
Affiliation(s)
- Sanda Mimouna
- The Institute of Research on Cancer and Aging, Nice, France
| | | | | | | | | | | |
Collapse
|
15
|
Kelly CJ, Colgan SP. Targeting Hypoxia to Augment Mucosal Barrier Function. JOURNAL OF EPITHELIAL BIOLOGY & PHARMACOLOGY 2012; 5:67-76. [PMID: 28824735 PMCID: PMC5560425 DOI: 10.2174/1875044301205010067] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sites of inflammation are associated with profound changes in tissue metabolism. Studies in vitro and in vivo have shown that the activation of the hypoxia-inducible factor (HIF) serves as an adaptive pathway for the resolution of inflammation associated with various murine disease models. The resolution of disease occurs, at least in part, through transcriptional regulation of non-classical epithelial barrier genes. There is significant recent interest in harnessing hypoxia-inducible pathways, including targeting the HIF and the proyl-hydroxylase (PHD) enzymes that stabilize HIF, to promote mucosal healing. Here, we review the signaling pathways involved and define how hypoxia-associated signaling provides mechanistic insight into augmenting barrier function in mucosal inflammatory disease.
Collapse
Affiliation(s)
- Caleb J Kelly
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Health Sciences Center, Aurora, CO USA
| | - Sean P Colgan
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Health Sciences Center, Aurora, CO USA
| |
Collapse
|
16
|
Castellanos-Rubio A, Caja S, Irastorza I, Fernandez-Jimenez N, Plaza-Izurieta L, Vitoria JC, Maki M, Lindfors K, Bilbao JR. Angiogenesis-related gene expression analysis in celiac disease. Autoimmunity 2012; 45:264-70. [PMID: 22136669 DOI: 10.3109/08916934.2011.637531] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Celiac disease (CD) involves disturbance of the small-bowel mucosal vascular network, and transglutaminase autoantibodies (TGA) have been related to angiogenesis disturbance, a complex phenomenon probably also influenced by common genetic variants in angiogenesis-related genes. A set of genes with "angiogenesis" GO term identified in a previous expression microarray experiment (SCG2, STAB1, TGFA, ANG, ERBB2, GNA13, PML, CASP8, ECGF1, JAG1, HIF1A, TNFSF13 and TGM2) was selected for genetic and functional studies. SNPs that showed a trend for association with CD in the first GWAS were genotyped in 555 patients and 541 controls. Gene expression of all genes was quantified in 15 pairs of intestinal biopsies (diagnosis vs. GFD) and in three-dimensional HUVEC and T84 cell cultures incubated with TGA-positive and negative serum. A regulatory SNP in TNFSF13 (rs11552708) is associated with CD (p = 0.01, OR = 0.7). Expression changes in biopsies pointed to TGM2 and PML as up-regulated antiangiogenic genes and to GNA13, TGFA, ERBB2 and SCG2 as down-regulated proangiogenic factors in CD. TGA seem to enhance TGM2 expression in both cell models, but PML expression was induced only in T84 enterocytes while GNA13 and ERBB2 were repressed in HUVEC endothelial cells, with several genes showing discordant effects in each model, highlighting the complexity of gene interactions in the pathogenesis of CD. Finally, cell culture models are useful tools to help dissect complex responses observed in human explants.
Collapse
Affiliation(s)
- Ainara Castellanos-Rubio
- Immunogenetics Research Laboratory, Endocrinology, Diabetes and Nutrition Research Group, Hospital de Cruces, Barakaldo, Basque Country
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Olson N, van der Vliet A. Interactions between nitric oxide and hypoxia-inducible factor signaling pathways in inflammatory disease. Nitric Oxide 2011; 25:125-37. [PMID: 21199675 DOI: 10.1016/j.niox.2010.12.010] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 12/22/2010] [Accepted: 12/29/2010] [Indexed: 02/06/2023]
Abstract
Induction and activation of nitric oxide (NO) synthases (NOS) and excessive production of NO are common features of almost all diseases associated with infection and acute or chronic inflammation, although the contribution of NO to the pathophysiology of these diseases is highly multifactorial and often still a matter of controversy. Because of its direct impact on tissue oxygenation and cellular oxygen (O(2)) consumption and re-distribution, the ability of NO to regulate various aspects of hypoxia-induced signaling has received widespread attention. Conditions of tissue hypoxia and the activation of hypoxia-inducible factors (HIF) have been implicated in hypoxia or in cancer biology, but are also being increasingly recognized as important features of acute and chronic inflammation. Thus, the activation of HIF transcription factors has been increasingly implicated in inflammatory diseases, and recent studies have indicated its critical importance in regulating phagocyte function, inflammatory mediator production, and regulation of epithelial integrity and repair processes. Finally, HIF also appears to contribute to important features of tissue fibrosis and epithelial-to-mesenchymal transition, processes that are associated with tissue remodeling in various non-malignant chronic inflammatory disorders. In this review, we briefly summarize the current state of knowledge with respect to the general mechanisms involved in HIF regulation and the impact of NO on HIF activation. Secondly, we will summarize the major recent findings demonstrating a role for HIF signaling in infection, inflammation, and tissue repair and remodeling, and will address the involvement of NO. The growing interest in hypoxia-induced signaling and its relation with NO biology is expected to lead to further insights into the complex roles of NO in acute or chronic inflammatory diseases and may point to the importance of HIF signaling as key feature of NO-mediated events during these disorders.
Collapse
Affiliation(s)
- Nels Olson
- Department of Pathology, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | | |
Collapse
|