Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39126102

ABSTRACT

Eosinophilic esophagitis (EoE) and inflammatory bowel disease (IBD) are chronic inflammatory disorders of the gastrointestinal tract, with EoE predominantly provoked by food and aeroallergens, whereas IBD is driven by a broader spectrum of immunopathological and environmental triggers. This review presents a comprehensive comparison of the pathophysiological and therapeutic strategies for EoE and IBD. We examine the current understanding of their underlying mechanisms, particularly the interplay between environmental factors and genetic susceptibility. A crucial element in both diseases is the integrity of the epithelial barrier, whose disruption plays a central role in their pathogenesis. The involvement of eosinophils, mast cells, B cells, T cells, dendritic cells, macrophages, and their associated cytokines is examined, highlighting the importance of targeting cytokine signaling pathways to modulate immune-epithelial interactions. We propose that advances in computation tools will uncover the significance of G-protein coupled receptors (GPCRs) in connecting immune and epithelial cells, leading to novel therapies for EoE and IBD.


Subject(s)
Eosinophilic Esophagitis , Inflammatory Bowel Diseases , Humans , Eosinophilic Esophagitis/etiology , Eosinophilic Esophagitis/immunology , Eosinophilic Esophagitis/pathology , Eosinophilic Esophagitis/therapy , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Animals , Cytokines/metabolism , Eosinophils/metabolism , Eosinophils/immunology , Eosinophils/pathology , Genetic Predisposition to Disease
2.
Gut ; 72(5): 821-833, 2023 05.
Article in English | MEDLINE | ID: mdl-35613844

ABSTRACT

OBJECTIVE: Disruption of the epithelial barrier plays an essential role in developing eosinophilic oesophagitis (EoE), a disease defined by type 2 helper T cell (Th2)-mediated food-associated and aeroallergen-associated chronic inflammation. Although an increased expression of interleukin (IL)-20 subfamily members, IL-19, IL-20 and IL-24, in Th2-mediated diseases has been reported, their function in EoE remains unknown. DESIGN: Combining transcriptomic, proteomic and functional analyses, we studied the importance of the IL-20 subfamily for EoE using patient-derived oesophageal three-dimensional models and an EoE mouse model. RESULTS: Patients with active EoE have increased expression of IL-20 subfamily cytokines in the oesophagus and serum. In patient-derived oesophageal organoids stimulated with IL-20 cytokines, RNA sequencing and mass spectrometry revealed a downregulation of genes and proteins forming the cornified envelope, including filaggrins. On the contrary, abrogation of IL-20 subfamily signalling in Il20R2 -/- animals resulted in attenuated experimental EoE reflected by reduced eosinophil infiltration, lower Th2 cytokine expression and preserved expression of filaggrins in the oesophagus. Mechanistically, these observations were mediated by the mitogen-activated protein kinase (MAPK); extracellular-signal regulated kinases (ERK)1/2) pathway. Its blockade prevented epithelial barrier impairment in patient-derived air-liquid interface cultures stimulated with IL-20 cytokines and attenuated experimental EoE in mice. CONCLUSION: Our findings reveal a previously unknown regulatory role of the IL-20 subfamily for oesophageal barrier function in the context of EoE. We propose that aberrant IL-20 subfamily signalling disturbs the oesophageal epithelial barrier integrity and promotes EoE development. Our study suggests that specific targeting of the IL-20 subfamily signalling pathway may present a novel strategy for the treatment of EoE.


Subject(s)
Eosinophilic Esophagitis , Animals , Mice , Cytokines/metabolism , Filaggrin Proteins , Interleukins/pharmacology , Interleukins/metabolism , Proteomics , Humans
3.
Gut ; 71(12): 2526-2538, 2022 12.
Article in English | MEDLINE | ID: mdl-35058274

ABSTRACT

OBJECTIVE: Mucosal-associated invariant T (MAIT) cells are the most abundant T cells in human liver. They respond to bacterial metabolites presented by major histocompatibility complex-like molecule MR1. MAIT cells exert regulatory and antimicrobial functions and are implicated in liver fibrogenesis. It is not well understood which liver cells function as antigen (Ag)-presenting cells for MAIT cells, and under which conditions stimulatory Ags reach the circulation. DESIGN: We used different types of primary human liver cells in Ag-presentation assays to blood-derived and liver-derived MAIT cells. We assessed MAIT cell stimulatory potential of serum from healthy subjects and patients with portal hypertension undergoing transjugular intrahepatic portosystemic shunt stent, and patients with inflammatory bowel disease (IBD). RESULTS: MAIT cells were dispersed throughout healthy human liver and all tested liver cell types stimulated MAIT cells, hepatocytes being most efficient. MAIT cell activation by liver cells occurred in response to bacterial lysate and pure Ag, and was prevented by non-activating MR1 ligands. Serum derived from peripheral and portal blood, and from patients with IBD stimulated MAIT cells in MR1-dependent manner. CONCLUSION: Our findings reveal previously unrecognised roles of liver cells in Ag metabolism and activation of MAIT cells, repression of which creates an opportunity to design antifibrotic therapies. The presence of MAIT cell stimulatory Ags in serum rationalises the observed activated MAIT cell phenotype in liver. Increased serum levels of gut-derived MAIT cell stimulatory ligands in patients with impaired intestinal barrier function indicate that intrahepatic Ag-presentation may represent an important step in the development of liver disease.


Subject(s)
Inflammatory Bowel Diseases , Mucosal-Associated Invariant T Cells , Humans , Minor Histocompatibility Antigens , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Liver/metabolism , Hepatocytes/metabolism , Inflammatory Bowel Diseases/metabolism , Lymphocyte Activation
4.
FASEB J ; 34(6): 7311-7329, 2020 06.
Article in English | MEDLINE | ID: mdl-32285992

ABSTRACT

Clinical studies suggest that pregnant women with elevated iron levels are more vulnerable to develop gestational diabetes mellitus (GDM), but the causes and underlying mechanisms are unknown. We hypothesized that hyperglycemia induces cellular stress responses leading to dysregulated placental iron homeostasis. Hence, we compared the expression of genes/proteins involved in iron homeostasis in placentae from GDM and healthy pregnancies (n = 11 each). RT-qPCR and LC-MS/MS analyses revealed differential regulation of iron transporters/receptors (DMT1/FPN1/ZIP8/TfR1), iron sensors (IRP1), iron regulators (HEPC), and iron oxidoreductases (HEPH/Zp). To identify the underlying mechanisms, we adapted BeWo trophoblast cells to normoglycemic (N), hyperglycemic (H), and hyperglycemic-hyperlipidemic (HL) conditions and assessed Fe3+ -uptake, expression patterns, and cellular pathways involving oxidative stress (OS), ER-stress, and autophagy. H and HL induced alterations in cellular morphology, differential iron transporter expression, and reduced Fe3+ -uptake confirming the impact of hyperglycemia on iron transport observed in GDM patients. Pathway analysis and rescue experiments indicated that dysregulated OS and disturbed autophagy processes contribute to the reduced placental iron transport under hyperglycemic conditions. These adaptations could represent a protective mechanism preventing the oxidative damage for both fetus and placenta caused by highly oxidative iron. In pregnancies with risk for GDM, antioxidant treatment, and controlled iron supplementation could help to balance placental OS levels protecting mother and fetus from impaired iron homeostasis.


Subject(s)
Diabetes, Gestational/metabolism , Diabetes, Gestational/physiopathology , Homeostasis/physiology , Iron/metabolism , Placenta/metabolism , Placenta/physiopathology , Adult , Antigens, CD/metabolism , Antioxidants/metabolism , Autophagy/physiology , Cation Transport Proteins/metabolism , Chromatography, Liquid/methods , Female , Ferritins/metabolism , Fetus/metabolism , Fetus/physiopathology , Humans , Male , Oxidative Stress/physiology , Pregnancy , Receptors, Transferrin/metabolism , Tandem Mass Spectrometry/methods , Trophoblasts/metabolism , Trophoblasts/physiology
5.
Int J Mol Sci ; 22(24)2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34948435

ABSTRACT

Maintaining intestinal health requires clear segregation between epithelial cells and luminal microbes. The intestinal mucus layer, produced by goblet cells (GCs), is a key element in maintaining the functional protection of the epithelium. The importance of the gut mucus barrier is highlighted in mice lacking Muc2, the major form of secreted mucins. These mice show closer bacterial residence to epithelial cells, develop spontaneous colitis and became moribund when infected with the attaching and effacing pathogen, Citrobacter rodentium. Furthermore, numerous observations have associated GCs and mucus layer dysfunction to the pathogenesis of inflammatory bowel disease (IBD). However, the molecular mechanisms that regulate the physiology of GCs and the mucus layer remain obscured. In this review, we consider novel findings describing divergent functionality and expression profiles of GCs subtypes within intestinal crypts. We also discuss internal (host) and external (diets and bacteria) factors that modulate different aspects of the mucus layer as well as the contribution of an altered mucus barrier to the onset of IBD.


Subject(s)
Epithelial Cells , Gastrointestinal Microbiome , Mucins/metabolism , Animals , Colitis , Goblet Cells/metabolism , Humans , Inflammatory Bowel Diseases , Mice , Mucins/physiology
6.
Carcinogenesis ; 38(12): 1157-1166, 2017 12 07.
Article in English | MEDLINE | ID: mdl-28968883

ABSTRACT

Evolution led to an essential symbiotic relationship between the host and commensal microbiota, regulating physiological functions including inflammation and immunity. This equilibrium can be disturbed by environmental factors such as lifestyle, diet or antibiotic pressure, contributing to create a dysbiosis. There is much evidence about the gut microbiota's contribution to carcinogenesis, involving pro-inflammatory and immunosuppressive signals. At the same time, it seems to be increasingly clear that commensal microbes can modulate cancer therapy efficacy and safety, in particular, innovating treatments as immune checkpoint inhibitors. In this review, we discuss how the microbiota can promote digestive tract carcinogenesis, responsiveness to cancer therapeutics and cancer-associated complications.


Subject(s)
Dysbiosis/complications , Gastrointestinal Microbiome , Gastrointestinal Neoplasms/microbiology , Animals , Humans
7.
FASEB J ; 30(10): 3598-3612, 2016 10.
Article in English | MEDLINE | ID: mdl-27435264

ABSTRACT

Deficiency in methyl donor (folate and vitamin B12) and in vitamin D is independently associated with altered bone development. Previously, methyl donor deficiency (MDD) was shown to weaken the activity of nuclear receptor coactivator, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α), for nuclear signaling in rat pups, including estrogen receptor-α and estrogen-related receptor-α; its effect on vitamin D receptor (VDR) signaling, however, is unknown. We studied bone development under MDD in rat pups and used human MG-63 preosteoblast cells to better understand the associated molecular mechanism. In young rats, MDD decreased total body bone mineral density, reduced tibia length, and impaired growth plate maturation, and in preosteoblasts, MDD slowed cellular proliferation. Mechanistic studies revealed decreased expression of VDR, estrogen receptor-α, PGC1α, arginine methyltransferase 1, and sirtuin 1 in both rat proximal diaphysis of femur and in MG-63, as well as decreased nuclear VDR-PGC1α interaction in MG-63 cells. The weaker VDR-PGC1α interaction could be attributed to the reduced protein expression, imbalanced PGC1α methylation/acetylation, and nuclear VDR sequestration by heat shock protein 90 (HSP90). These together compromised bone development, which is reflected by lowered bone alkaline phosphatase and increased proadipogenic peroxisome proliferator-activated receptor-γ, adiponectin, and estrogen-related receptor-α expression. Of interest, under MDD, the bone development effects of 1,25-dihydroxyvitamin D3 were ineffectual and these could be rescued by the addition of S-adenosylmethionine, which restored expression of arginine methyltransferase 1, PGC1α, adiponectin, and HSP90. In conclusion, MDD inactivates vitamin D signaling via both disruption of VDR-PGC1α interaction and sequestration of nuclear VDR attributable to HSP90 overexpression. These data suggest that vitamin D treatment may be ineffective under MDD.-Feigerlova, E., Demarquet, L., Melhem, H., Ghemrawi, R., Battaglia-Hsu, S.-F., Ewu, E., Alberto, J.-M., Helle, D., Weryha, G., Guéant, J.-L. Methyl donor deficiency impairs bone development via peroxisome proliferator-activated receptor-γ coactivator-1α-dependent vitamin D receptor pathway.


Subject(s)
Bone Development/physiology , PPAR gamma/metabolism , Receptors, Calcitriol/metabolism , Animals , Calcitriol/metabolism , Cell Line, Tumor , Female , Heat-Shock Proteins/metabolism , Humans , Rats , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , ERRalpha Estrogen-Related Receptor
8.
Gut ; 65(4): 595-606, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25608526

ABSTRACT

BACKGROUND: Methyl donor deficiency (MDD) aggravates experimental colitis in rats and increases endoplasmic reticulum (ER) stress through decreased sirtuin 1 (SIRT1) in neuronal cells and myocardium. ER stress plays a key role in IBD pathogenesis. AIM: We investigated whether the influence of MDD on colitis resulted from an ER stress response triggered by decreased SIRT1 expression. DESIGN: The unfolded protein response (UPR), chaperones proteins, heat shock factor protein 1 (HSF1) and SIRT1 were examined in rats with MDD and dextran sulfate sodium (DSS)-induced colitis in a Caco-2 cell model with stable expression of transcobalamin-oleosin (TO) chimera, which impairs cellular availability of vitamin B12, and in IBD. The effects of SIRT1 activation were studied both in vitro and in vivo. RESULTS: MDD aggravated DSS-induced colitis clinically, endoscopically and histologically. MDD activated ER stress pathways, with increased phosphorylate-PKR-like ER kinase, P-eiF-2α, P-IRE-1α, activating transcription factor (ATF)6, XBP1-S protein and ATF4 mRNA expression levels in rats. This was accompanied by reduced SIRT1 expression level and greater acetylation of HSF1, in relation with a dramatic decrease of chaperones (binding immunoglobulin protein (BIP), heat shock protein (HSP)27 and HSP90). Adding either vitamin B12, S-adenosylmethionine or an SIRT1 activator (SRT1720) reduced the UPR in vitro. In rats, SIRT1 activation by SRT1720 prevented colitis by reducing HSF1 acetylation and increasing expression of BIP, HSP27 and HSP90. Immunohistochemistry showed impaired expression of SIRT1 in the colonic epithelium of patients with IBD. CONCLUSIONS: SIRT1 is a master regulator of ER stress and severity of experimental colitis in case of MDD. It could deserve further interest as a therapeutic target of IBD.


Subject(s)
Biopsy , Colitis/chemically induced , Diet , Endoplasmic Reticulum Stress , Sirtuin 1/metabolism , Animals , Blotting, Western , Caco-2 Cells , Cells, Cultured , Choline Deficiency , DNA-Binding Proteins , Dextran Sulfate/pharmacology , Eukaryotic Initiation Factor-2/metabolism , Female , Folic Acid Deficiency , Humans , Immunoenzyme Techniques , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Transcription Factors , Transfection , Unfolded Protein Response , Vitamin B 12 Deficiency , eIF-2 Kinase
9.
J Plast Reconstr Aesthet Surg ; 87: 352-360, 2023 12.
Article in English | MEDLINE | ID: mdl-37925927

ABSTRACT

OBJECTIVE: Hepatic artery anastomosis in liver transplantations requires a meticulous technique to minimize the risk of hepatic artery thrombosis (HAT). The microscope helped improve anastomosis techniques in pediatric patients with small caliber vessels. The aim of this review was to compare microsurgical and non-microsurgical techniques on the incidence of HAT in liver transplantations. The secondary objective was to compare HAT incidence between pediatric and adult cohorts and between plastic and transplant surgeons. METHODS: A systematic review of the literature using Medline, Embase, and Cumulative Index to Nursing and Allied Health Literature (CINAHL) was conducted on studies involving HAT in liver transplantations with microsurgery. Three reviewers performed a full article review and data extraction for studies meeting the eligibility criteria of the study. RESULTS: Forty-five studies were incorporated in the final analysis. A total of 7346 patients and 7506 liver transplants were included. The mean age was 17 years old with an equivalent distribution between pediatric (51%, n = 3218) and adult patients (49%, n = 3145). A total of 6351 of these transplantations underwent microsurgical repair, against 1157 with non-microsurgical techniques. The overall HAT rate was 4.9%, including 4.2% in the microsurgical group (n = 268) and 8.5% in the non-microsurgical group (n = 98), a statistically significant increase of 4.3%. The occurrence of HAT was 2.6% with a plastic surgeon versus 4.6% with other types of surgeons. When using microsurgical techniques, the HAT rate was 4.2% with living donors versus 7.7% with deceased donors. CONCLUSIONS: HAT and subsequent liver transplant failure are lower when microsurgical techniques, living donors, and plastic surgeons with a microsurgical training are involved in the operation.


Subject(s)
Liver Transplantation , Thrombosis , Adult , Humans , Child , Adolescent , Liver Transplantation/adverse effects , Liver Transplantation/methods , Hepatic Artery/surgery , Anastomosis, Surgical/methods , Thrombosis/epidemiology , Thrombosis/etiology , Thrombosis/surgery , Microsurgery/methods , Living Donors , Retrospective Studies
10.
Cell Rep ; 42(8): 113005, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37590143

ABSTRACT

The intricate interplay between gut microbes and the onset of experimental autoimmune encephalomyelitis (EAE) remains poorly understood. Here, we uncover remarkable similarities between CD4+ T cells in the spinal cord and their counterparts in the small intestine. Furthermore, we unveil a synergistic relationship between the microbiota, particularly enriched with the tryptophan metabolism gene EC:1.13.11.11, and intestinal cells. This symbiotic collaboration results in the biosynthesis of kynurenic acid (KYNA), which modulates the recruitment and aggregation of GPR35-positive macrophages. Subsequently, a robust T helper 17 (Th17) immune response is activated, ultimately triggering the onset of EAE. Conversely, modulating the KYNA-mediated GPR35 signaling in Cx3cr1+ macrophages leads to a remarkable amelioration of EAE. These findings shed light on the crucial role of microbial-derived tryptophan metabolites in regulating immune responses within extraintestinal tissues.


Subject(s)
Encephalitis , Encephalomyelitis, Autoimmune, Experimental , Gastrointestinal Microbiome , Animals , Kynurenic Acid , Tryptophan , Macrophages
11.
Mucosal Immunol ; 15(3): 443-458, 2022 03.
Article in English | MEDLINE | ID: mdl-35264769

ABSTRACT

Goblet cells secrete mucin to create a protective mucus layer against invasive bacterial infection and are therefore essential for maintaining intestinal health. However, the molecular pathways that regulate goblet cell function remain largely unknown. Although GPR35 is highly expressed in colonic epithelial cells, its importance in promoting the epithelial barrier is unclear. In this study, we show that epithelial Gpr35 plays a critical role in goblet cell function. In mice, cell-type-specific deletion of Gpr35 in epithelial cells but not in macrophages results in goblet cell depletion and dysbiosis, rendering these animals more susceptible to Citrobacter rodentium infection. Mechanistically, scRNA-seq analysis indicates that signaling of epithelial Gpr35 is essential to maintain normal pyroptosis levels in goblet cells. Our work shows that the epithelial presence of Gpr35 is a critical element for the function of goblet cell-mediated symbiosis between host and microbiota.


Subject(s)
Enterobacteriaceae Infections , Goblet Cells , Animals , Citrobacter rodentium , Colon/microbiology , Enterobacteriaceae Infections/metabolism , Goblet Cells/physiology , Intestinal Mucosa/metabolism , Mice , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
12.
Front Immunol ; 12: 717392, 2021.
Article in English | MEDLINE | ID: mdl-34790192

ABSTRACT

Diet and gut microbial metabolites mediate host immune responses and are central to the maintenance of intestinal health. The metabolite-sensing G-protein coupled receptors (GPCRs) bind metabolites and trigger signals that are important for the host cell function, survival, proliferation and expansion. On the contrary, inadequate signaling of these metabolite-sensing GPCRs most likely participate to the development of diseases including inflammatory bowel diseases (IBD). In the intestine, metabolite-sensing GPCRs are highly expressed by epithelial cells and by specific subsets of immune cells. Such receptors provide an important link between immune system, gut microbiota and metabolic system. Member of these receptors, GPR35, a class A rhodopsin-like GPCR, has been shown to be activated by the metabolites tryptophan-derived kynurenic acid (KYNA), the chemokine CXCL17 and phospholipid derivate lysophosphatidic acid (LPA) species. There have been studies on GPR35 in the context of intestinal diseases since its identification as a risk gene for IBD. In this review, we discuss the pharmacology of GPR35 including its proposed endogenous and synthetic ligands as well as its antagonists. We elaborate on the risk variants of GPR35 implicated in gut-related diseases and the mechanisms by which GPR35 contribute to intestinal homeostasis.


Subject(s)
Homeostasis , Inflammatory Bowel Diseases/pathology , Intestines/physiology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Chemokines, CXC/metabolism , Humans , Kynurenic Acid/metabolism , Lysophospholipids/metabolism
13.
Sci Rep ; 10(1): 1438, 2020 01 29.
Article in English | MEDLINE | ID: mdl-31996710

ABSTRACT

Proton-sensing ovarian cancer G-protein coupled receptor (OGR1) plays an important role in pH homeostasis. Acidosis occurs at sites of intestinal inflammation and can induce endoplasmic reticulum (ER) stress and the unfolded protein response (UPR), an evolutionary mechanism that enables cells to cope with stressful conditions. ER stress activates autophagy, and both play important roles in gut homeostasis and contribute to the pathogenesis of inflammatory bowel disease (IBD). Using a human intestinal epithelial cell model, we investigated whether our previously observed protective effects of OGR1 deficiency in experimental colitis are associated with a differential regulation of ER stress, the UPR and autophagy. Caco-2 cells stably overexpressing OGR1 were subjected to an acidic pH shift. pH-dependent OGR1-mediated signalling led to a significant upregulation in the ER stress markers, binding immunoglobulin protein (BiP) and phospho-inositol required 1α (IRE1α), which was reversed by a novel OGR1 inhibitor and a c-Jun N-terminal kinase (JNK) inhibitor. Proton-activated OGR1-mediated signalling failed to induce apoptosis, but triggered accumulation of total microtubule-associated protein 1 A/1B-light chain 3, suggesting blockage of late stage autophagy. Our results show novel functions for OGR1 in the regulation of ER stress through the IRE1α-JNK signalling pathway, as well as blockage of autophagosomal degradation. OGR1 inhibition might represent a novel therapeutic approach in IBD.


Subject(s)
Endoribonucleases/metabolism , Epithelial Cells/metabolism , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/metabolism , Microtubules/metabolism , Ovarian Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Acidosis , Autophagy , Caco-2 Cells , Endoplasmic Reticulum Stress/genetics , Female , Homeostasis , Humans , Hydrogen-Ion Concentration , MAP Kinase Signaling System , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Unfolded Protein Response
14.
Commun Biol ; 3(1): 130, 2020 03 18.
Article in English | MEDLINE | ID: mdl-32188932

ABSTRACT

Comprehensive development is critical for gut macrophages being essential for the intestinal immune system. However, the underlying mechanisms of macrophage development in the colon remain elusive. To investigate the function of branched-chain amino acids in the development of gut macrophages, an inducible knock-out mouse model for the branched-chain amino acid transporter CD98hc in CX3CR1+ macrophages was generated. The relatively selective deletion of CD98hc in macrophage populations leads to attenuated severity of chemically-induced colitis that we assessed by clinical, endoscopic, and histological scoring. Single-cell RNA sequencing of colonic lamina propria macrophages revealed that conditional deletion of CD98hc alters the "monocyte waterfall"-development to MHC II+ macrophages. The change in the macrophage development after deletion of CD98hc is associated with increased apoptotic gene expression. Our results show that CD98hc deletion changes the development of colonic macrophages.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Colitis/metabolism , Colon/metabolism , Fusion Regulatory Protein 1, Heavy Chain/deficiency , Intestinal Mucosa/metabolism , Macrophages/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Colitis/chemically induced , Colitis/pathology , Colitis/prevention & control , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Colon/ultrastructure , Crohn Disease/diagnosis , Crohn Disease/genetics , Crohn Disease/metabolism , Dextran Sulfate , Disease Models, Animal , Female , Fusion Regulatory Protein 1, Heavy Chain/genetics , Gene Expression Regulation , Humans , Intestinal Mucosa/ultrastructure , Macrophages/ultrastructure , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Phenotype , RNA-Seq , Single-Cell Analysis , Young Adult
15.
Cell Rep ; 32(5): 107979, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32755573

ABSTRACT

Single-nucleotide polymorphisms in the gene encoding G protein-coupled receptor 35 (GPR35) are associated with increased risk of inflammatory bowel disease. However, the mechanisms by which GPR35 modulates intestinal immune homeostasis remain undefined. Here, integrating zebrafish and mouse experimental models, we demonstrate that intestinal Gpr35 expression is microbiota dependent and enhanced upon inflammation. Moreover, murine GPR35+ colonic macrophages are characterized by enhanced production of pro-inflammatory cytokines. We identify lysophosphatidic acid (LPA) as a potential endogenous ligand produced during intestinal inflammation, acting through GPR35 to induce tumor necrosis factor (Tnf) expression in macrophages. Mice lacking Gpr35 in CX3CR1+ macrophages aggravate colitis when exposed to dextran sodium sulfate, which is associated with decreased transcript levels of the corticosterone-generating gene Cyp11b1 and macrophage-derived Tnf. Administration of TNF in these mice restores Cyp11b1 expression and intestinal corticosterone production and ameliorates DSS-induced colitis. Our findings indicate that LPA signals through GPR35 in CX3CR1+ macrophages to maintain TNF-mediated intestinal homeostasis.


Subject(s)
CX3C Chemokine Receptor 1/metabolism , Homeostasis , Intestines/physiology , Lysophospholipids/metabolism , Macrophages/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Zebrafish Proteins/metabolism , Animals , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate , Gastrointestinal Microbiome , Gene Deletion , Humans , Inflammation/pathology , Inflammatory Bowel Diseases/pathology , Mice, Inbred C57BL , Phosphoric Diester Hydrolases/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Zebrafish
16.
Cells ; 8(5)2019 05 14.
Article in English | MEDLINE | ID: mdl-31091682

ABSTRACT

Increasing evidence has indicated that diet and metabolites, including bacteria- and host-derived metabolites, orchestrate host pathophysiology by regulating metabolism, immune system and inflammation. Indeed, autoimmune diseases such as inflammatory bowel disease (IBD) are associated with the modulation of host response to diets. One crucial mechanism by which the microbiota affects the host is signaling through G protein-coupled receptors (GPCRs) termed metabolite-sensing GPCRs. In the gut, both immune and nonimmune cells express GPCRs and their activation generally provide anti-inflammatory signals through regulation of both the immune system functions and the epithelial integrity. Members of GPCR family serve as a link between microbiota, immune system and intestinal epithelium by which all these components crucially participate to maintain the gut homeostasis. Conversely, impaired GPCR signaling is associated with IBD and other diseases, including hepatic steatosis, diabetes, cardiovascular disease, and asthma. In this review, we first outline the signaling, function, expression and the physiological role of several groups of metabolite-sensing GPCRs. We then discuss recent findings on their role in the regulation of the inflammation, their existing endogenous and synthetic ligands and innovative approaches to therapeutically target inflammatory bowel disease.


Subject(s)
Autoimmune Diseases , Bacteria/metabolism , Gastrointestinal Microbiome/immunology , Inflammation , Inflammatory Bowel Diseases , Receptors, G-Protein-Coupled/physiology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/microbiology , Diet , Homeostasis , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Mice
17.
Sci Rep ; 9(1): 6225, 2019 04 17.
Article in English | MEDLINE | ID: mdl-30996342

ABSTRACT

High levels of atherogenic lipids in pregnancy are associated with health complications for the mother, the fetus and the newborn. As endocrine secretory tissue, the human placenta releases apolipoproteins (apos), particularly apoA1 and apoE. However, the magnitude and the directionality of the apo secretions remain unknown. We aimed to 1) determine the amount and orientation (apical-maternal versus basal-fetal) of placentally secreted apoA1 and apoE using human perfused placenta and primary trophoblast cell (PTC) culture, 2) compare apoA1 and apoE secretions of PTC with that of hepatocytes and 3) associate the obtained results with human blood levels by determining apoA1 and apoE concentrations in maternal and fetal serum samples. In perfused placenta and serum samples, apoA1 and apoE concentrations were significantly higher at the maternal compared to the fetal side. For apoE a similar trend was found in PTC. For apoA1, the secretion to the apical side declined over time while release to the basal side was stable resulting in significantly different apoA1 concentrations between both sides. Unexpectedly, PTC secreted significantly higher amounts of apoA1 and apoE compared to hepatocytes. Our data indicate that the placenta may play an important role in maternal and fetal cholesterol homeostasis via secretion of anti-atherogenic apos.


Subject(s)
Apolipoprotein A-I/blood , Apolipoproteins E/blood , Atherosclerosis/metabolism , Cholesterol/metabolism , Fetus/metabolism , Homeostasis/physiology , Trophoblasts/metabolism , Adult , Biological Transport/physiology , Cells, Cultured , Female , Hepatocytes/metabolism , Humans , Pregnancy
18.
J Crohns Colitis ; 13(6): 785-797, 2019 May 27.
Article in English | MEDLINE | ID: mdl-30590526

ABSTRACT

BACKGROUND AND AIMS: Inflammatory bowel disease [IBD] is accompanied by lesions in the epithelial barrier, which allow translocation of bacterial products from the gut lumen to the host's circulation. IMM-124E is a colostrum-based product containing high levels of anti-E.coli-LPS IgG, and might limit exposure to bacterial endotoxins. Here, we investigated whether IMM-124E can ameliorate intestinal inflammation. METHODS: Acute colitis was induced in WT C57Bl/6J mice by administration of 2.5% dextran sodium sulphate [DSS] for 7 days. T cell transfer colitis was induced via transfer of 0.5 x 106 naïve T cells into RAG2-/- C57Bl/6J mice. IMM-124E was administered daily by oral gavage, either preventively or therapeutically. RESULTS: Treatment with IMM-124E significantly ameliorated colitis in acute DSS colitis and in T cell transfer colitis. Maximum anti-inflammatory effects were detected at an IMM-124E concentration of 100 mg/kg body weight, whereas 25 mg/kg and 500 mg/kg were less effective. Histology revealed reduced levels of infiltrating immune cells and less pronounced mucosal damage. Flow cytometry revealed reduced numbers of effector T helper cells in the intestine, whereas levels of regulatory T cells were enhanced. IMM-124E treatment reduced the DSS-induced increase of serum levels of lipopolysaccharide [LPS]-binding protein, indicating reduced systemic LPS exposure. CONCLUSIONS: Our results demonstrate that oral treatment with IMM-124E significantly reduces intestinal inflammation, via decreasing the accumulation of pathogenic T cells and concomitantly increasing the induction of regulatory T cells. Our study confirms the therapeutic efficacy of IMM-124E in acute colitis and suggests that administration of IMM-124E might represent a novel therapeutic strategy to induce or maintain remission in chronic colitis.


Subject(s)
Colitis/drug therapy , Colostrum/chemistry , Animals , Blotting, Western , Cattle , Colitis/pathology , Colon/pathology , Colostrum/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunoglobulin G/immunology , Immunoglobulin G/therapeutic use , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Peroxidase/metabolism
19.
Cell Mol Gastroenterol Hepatol ; 7(2): 339-355, 2019.
Article in English | MEDLINE | ID: mdl-30704983

ABSTRACT

BACKGROUND & AIMS: Hypoxia-associated pathways influence the development of inflammatory bowel disease. Adaptive responses to hypoxia are mediated through hypoxia-inducible factors, which are regulated by iron-dependent hydroxylases. Signals reflecting oxygen tension and iron levels in enterocytes regulate iron metabolism. Conversely, iron availability modulates responses to hypoxia. In the present study we sought to elucidate how iron influences the responses to hypoxia in the intestinal epithelium. METHODS: Human subjects were exposed to hypoxia, and colonic biopsy specimens and serum samples were collected. HT-29, Caco-2, and T84 cells were subjected to normoxia or hypoxia in the presence of iron or the iron chelator deferoxamine. Changes in inflammatory gene expression and signaling were assessed by quantitative polymerase chain reaction and Western blot. Chromatin immunoprecipitation was performed using antibodies against nuclear factor (NF)-κB and primers for the promoter of tumor necrosis factor (TNF) and interleukin (IL)1ß. RESULTS: Human subjects presented reduced levels of ferritin in the intestinal epithelium after hypoxia. Hypoxia reduced iron deprivation-associated TNF and IL1ß expression in HT-29 cells through the induction of autophagy. Contrarily, hypoxia triggered TNF and IL1ß expression, and NF-κB activation in Caco-2 and T84 cells. Iron blocked autophagy in Caco-2 cells, while reducing hypoxia-associated TNF and IL1ß expression through the inhibition of NF-κB binding to the promoter of TNF and IL1ß. CONCLUSIONS: Hypoxia promotes iron mobilization from the intestinal epithelium. Hypoxia-associated autophagy reduces inflammatory processes in HT-29 cells. In Caco-2 cells, iron uptake is essential to counteract hypoxia-induced inflammation. Iron mobilization into enterocytes may be a vital protective mechanism in the hypoxic inflamed mucosa.


Subject(s)
Hypoxia/complications , Inflammation/drug therapy , Inflammation/etiology , Intestinal Mucosa/metabolism , Iron/therapeutic use , NF-kappa B/metabolism , Adult , Aged , Aged, 80 and over , Autophagy/drug effects , Caco-2 Cells , HT29 Cells , Humans , Inflammation/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Intestinal Mucosa/drug effects , Middle Aged , Models, Biological , Promoter Regions, Genetic/genetics , RNA Stability/drug effects , RNA Stability/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Young Adult
20.
PLoS One ; 13(2): e0193003, 2018.
Article in English | MEDLINE | ID: mdl-29447283

ABSTRACT

Gp96 is an endoplasmic reticulum chaperone for multiple protein substrates. Its lack in intestinal macrophages of Crohn's disease (CD) patients is correlated with loss of tolerance against the host gut flora. Gp96 has been stablished to be an essential chaperone for Toll-like receptors (TLRs). We studied the impact of gp96-knockdown on TLR-function in macrophages. TLR2 and TLR4 expression was only decreased but not abolished when gp96 was knocked-down in cell lines, whereas in a monocyte/macrophage specific knock-out mouse model (LysMCre) TLR4 was abolished, while TLR2 was still present. Lipopolysaccharide (LPS)-induced NF-κB activation was still observed in the absence of gp96, and gp96-deficient macrophages were able to up-regulate surface TLR4 upon LPS treatment, suggesting that there is another chaperone involved in the folding of TLR4 upon stress responses. Moreover, LPS-dependent pro-inflammatory cytokines were still expressed, although to a lesser extent in the absence of gp96, which reinforces the fact that gp96 is involved in regulating signaling cascades downstream of TLR4 are impaired upon loss of gp96. In addition, we have also found a reduced phosphorylation of ERK and p38 kinases and an impaired response upon CSF1R activation in gp96 deficient macrophages. Our findings indicate that the loss of gp96 not only impairs TLR4 signaling, but is also associated with a diminished phosphorylation of ERK and mitogen-activated stress kinases resulting in an impaired signalling through several receptors, including CSF1R.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Membrane Glycoproteins/deficiency , Toll-Like Receptor 4/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , HEK293 Cells , Humans , Interleukin-8/metabolism , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Phosphorylation/physiology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics
SELECTION OF CITATIONS
SEARCH DETAIL