Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
PLoS One ; 15(8): e0236657, 2020.
Article in English | MEDLINE | ID: mdl-32760089

ABSTRACT

Crohn's disease is a pathological condition of the gastro-intestinal tract, causing severe transmural inflammation in the ileum and/or colon. Cigarette smoking is one of the best known environmental risk factors for the development of Crohn's disease. Nevertheless, very little is known about the effect of prolonged cigarette smoke exposure on inflammatory modulators in the gut. We examined the effect of cigarette smoke on cytokine profiles in the healthy and inflamed gut of human subjects and in the trinitrobenzene sulphonic acid mouse model, which mimics distal Crohn-like colitis. In addition, the effect of cigarette smoke on epithelial expression of transient receptor potential channels and their concurrent increase with cigarette smoke-augmented cytokine production was investigated. Active smoking was associated with increased IL-8 transcription in ileum of controls (p < 0,001; n = 18-20/group). In the ileum, TRPV1 mRNA levels were decreased in never smoking Crohn's disease patients compared to healthy subjects (p <0,001; n = 20/group). In the colon, TRPV1 mRNA levels were decreased (p = 0,046) in smoking healthy controls (n = 20/group). Likewise, healthy mice chronically exposed to cigarette smoke (n = 10/group) showed elevated ileal Cxcl2 (p = 0,0075) and colonic Kc mRNA levels (p = 0,0186), whereas TRPV1 mRNA and protein levels were elevated in the ileum (p = 0,0315). Although cigarette smoke exposure prior to trinitrobenzene sulphonic acid administration did not alter disease activity, increased pro-inflammatory cytokine production was observed in the distal colon (Kc: p = 0,0273; Cxcl2: p = 0,104; Il1-ß: p = 0,0796), in parallel with the increase of Trpv1 mRNA (p < 0,001). We infer that CS affects pro-inflammatory cytokine expression in healthy and inflamed gut, and that the simultaneous modulation of TRPV1 may point to a potential involvement of TRPV1 in cigarette smoke-induced production of inflammatory mediators.


Subject(s)
Colon/metabolism , Crohn Disease/metabolism , Ileum/metabolism , TRPV Cation Channels/metabolism , Tobacco Smoking/adverse effects , Adult , Aged , Animals , Caco-2 Cells , Colon/pathology , Crohn Disease/chemically induced , Crohn Disease/pathology , Cytokines/metabolism , Disease Models, Animal , Female , HT29 Cells , Humans , Ileum/pathology , Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Translational Research, Biomedical , Trinitrobenzenesulfonic Acid
2.
Article in English | MEDLINE | ID: mdl-31969985

ABSTRACT

BACKGROUND: Brachyspira hyodysenteriae is the primary cause of swine dysentery, characterized by bloody to mucoid diarrhea due to mucohaemorhagic colitis in pigs and primarily affects pigs during the grow/finishing stage. Control and prevention of B. hyodysenteriae consists of administration of antimicrobial drugs, besides management and adapted feeding strategies. A worldwide re-emergence of the disease has recently been reported with an increasing number of isolates demonstrating decreased susceptibility to several crucially important antimicrobials in the control of swine dysentery. A novel non-antibiotic zinc chelate has been reported to demonstrate positive effects on fecal quality and consistency, general clinical signs, average daily weight gain and B. hyodysenteriae excretion during and after a 6-day oral treatment. The objective of the present study was to evaluate the zinc chelate (Intra Dysovinol® 499 mg/ml (ID); Elanco) on naturally occurring swine dysentery due to B. hyodysenteriae under field conditions in the Netherlands. RESULTS: Oral administration of zinc chelate resulted in improvement of general clinical signs from 3 days onwards in the ID-treated group combined with a significantly better total fecal score at 14 days post-treatment. Overall, average daily weight gain was better in the ID-treated group over the entire study period (0-14 days) and during the 8 days following the end of ID-treatment. A significant reduction (4.48 vs. 0.63 log10 cfu/g feces; ID-treated vs. control) in B. hyodysenteriae excretion was observed during the 6-day treatment period with a high percentage of animals (58.3 vs. 12.3%; ID-treated vs. control) with no excretion of B. hyodysenteriae from their feces. No additional antimicrobial treatment was needed in the ID-treated group, whereas 35% of the pigs in the control group were treated with an antibiotic at least once. No mortality occurred in both groups. No adverse events were reported during and following the ID-treatment. CONCLUSIONS: Zinc chelate - administered as a Zn-Na2-EDTA complex - is a non-antibiotic treatment for swine dysentery that reduces B. hyodysenteriae shedding with 4.48 log10 cfu/g feces within its 6-day treatment while improving general clinical signs (90.0 vs. 73.6% animals with normal score) and total fecal score within 2-4 days following administration in naturally infected pigs. The positive effects of ID treatment remain for at least 8 days after cessation of oral ID therapy. Pigs remaining in a highly contaminated environment may be re-infected following the end of ID treatment, however, this is not different to standard antimicrobial therapy. Therefore, control of swine dysentery should combine an efficacious treatment with additional management practices to reduce the environmental infection pressure in order to limit re-infection as much as possible. The ID treatment resulted in a higher growth rate and improved general health, whereas no mortality was observed and no additional therapeutic treatments were necessary in contrast to the control pigs.

3.
Pathobiology ; 84(1): 1-15, 2017.
Article in English | MEDLINE | ID: mdl-27388890

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by severe gastrointestinal inflammation and results from a complex interplay between genetic and environmental factors. IBD includes two prominent subtypes: Crohn's disease (CD) and ulcerative colitis (UC). One of the main risk factors for the development of CD is cigarette smoking, while UC is rather a disease of ex-smokers. To date, many of the mechanisms underlying the immune imbalance in IBD and the involvement of cigarette smoke (CS) are incompletely understood. Transient receptor potential (TRP) proteins are non-selective cation channels that, upon activation, lead to plasma membrane depolarization and, in general, to Ca2+ influx. TRP channels of the ankyrin and vanilloid family, expressed by sensory neurons in the central and enteric nervous systems, have been extensively studied in the context of intestinal inflammation. Moreover, recent advances made on the role of non-neuronal expressed TRP channels shed light on the involvement of epithelial cells in inflammatory processes. This review focuses on how CS may impact TRP channel function in intestinal inflammation. Firstly, we discuss the current knowledge on neuronal TRP channels, known to be linked to IBD, in health, immune homeostasis and intestinal inflammation. Subsequently, we address how TRP channels are activated by CS and its components in other organ systems and also hypothesize on the potential implications for CS-mediated TRP channel activation in gut inflammation.


Subject(s)
Inflammatory Bowel Diseases/etiology , Smoking/adverse effects , Transient Receptor Potential Channels/physiology , Colitis, Ulcerative/etiology , Colitis, Ulcerative/physiopathology , Crohn Disease/etiology , Crohn Disease/physiopathology , Epithelial Cells/pathology , Humans , Inflammation/etiology , Inflammation/physiopathology , Inflammatory Bowel Diseases/physiopathology
4.
Environ Microbiol ; 18(5): 1352-63, 2016 05.
Article in English | MEDLINE | ID: mdl-26033517

ABSTRACT

Inflammatory bowel diseases (IBD) are complex multifactorial diseases characterized by an inappropriate host response to an altered commensal microbiome and dysfunctional mucus barrier. Cigarette smoking is the best known environmental risk factor in IBD. Here, we studied the influence of chronic smoke exposure on the gut microbiome, mucus layer composition and immune factors in conventional mice. We compared smoke-exposed with air-exposed mice (n = 12) after a smoke exposure of 24 weeks. Both Illumina sequencing (n = 6) and denaturing gradient gel electrophoresis (n = 12) showed that bacterial activity and community structure were significantly altered in the colon due to smoke exposure. Interestingly, an increase of Lachnospiraceae sp. activity in the colon was observed. Also, the mRNA expression of Muc2 and Muc3 increased in the ileum, whereas Muc4 increased in the distal colon of smoke-exposed mice (n = 6). Furthermore, we observed increased Cxcl2 and decreased Ifn-γ in the ileum, and increased Il-6 and decreased Tgf-ß in the proximal colon. Tight junction gene expression remained unchanged. We infer that the modulating role of chronic smoke exposure as a latently present risk factor in the gut may be driven by the altered epithelial mucus profiles and changes in microbiome composition and immune factors.


Subject(s)
Gastrointestinal Microbiome , Inflammation Mediators/metabolism , Mucins/metabolism , Smoking , Animals , Bacteria/isolation & purification , Colon/metabolism , Colon/microbiology , Environmental Exposure , Gastrointestinal Tract/microbiology , Gene Expression , Ileum/metabolism , Male , Mice, Inbred C57BL , Mucins/genetics , Tobacco Products
5.
PLoS One ; 10(11): e0141570, 2015.
Article in English | MEDLINE | ID: mdl-26523550

ABSTRACT

The inflammatory cytokine TNF-α is a central mediator in many immune-mediated diseases, such as Crohn's disease (CD), spondyloarthritis (SpA) and chronic obstructive pulmonary disease (COPD). Epidemiologic studies have shown that cigarette smoking (CS) is a prominent common risk factor in these TNF-dependent diseases. We exposed TNFΔARE mice; in which a systemic TNF-α overexpression leads to the development of inflammation; to 2 or 4 weeks of air or CS. We investigated the effect of deregulated TNF expression on CS-induced pulmonary inflammation and the effect of CS exposure on the initiation and progression of gut and joint inflammation. Upon 2 weeks of CS exposure, inflammation in lungs of TNFΔARE mice was significantly aggravated. However, upon 4 weeks of CS-exposure, this aggravation was no longer observed. TNFΔARE mice have no increases in CD4+ and CD8+ T cells and a diminished neutrophil response in the lungs after 4 weeks of CS exposure. In the gut and joints of TNFΔARE mice, 2 or 4 weeks of CS exposure did not modulate the development of inflammation. In conclusion, CS exposure does not modulate gut and joint inflammation in TNFΔARE mice. The lung responses towards CS in TNFΔARE mice however depend on the duration of CS exposure.


Subject(s)
Arthritis/pathology , Inflammatory Bowel Diseases/pathology , Pneumonia/pathology , Smoking/adverse effects , Tumor Necrosis Factor-alpha/genetics , Acute-Phase Proteins/metabolism , Animals , Arthritis/genetics , Arthritis/immunology , Cytokines/blood , Disease Models, Animal , Feces/chemistry , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Lipocalin-2 , Lipocalins/metabolism , Mice , Oncogene Proteins/metabolism , Pneumonia/genetics , Pneumonia/immunology , Sequence Deletion , Tumor Necrosis Factor-alpha/metabolism
6.
EMBO J ; 34(4): 466-74, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25599993

ABSTRACT

Antinuclear antibodies are a hallmark feature of generalized autoimmune diseases, including systemic lupus erythematosus and systemic sclerosis. However, the processes underlying the loss of tolerance against nuclear self-constituents remain largely unresolved. Using mice deficient in lymphotoxin and Hox11, we report that approximately 25% of mice lacking secondary lymphoid organs spontaneously develop specific antinuclear antibodies. Interestingly, we find this phenotype is not caused by a defect in central tolerance. Rather, cell-specific deletion and in vivo lymphotoxin blockade link these systemic autoimmune responses to the formation of gut-associated lymphoid tissue in the neonatal period of life. We further demonstrate antinuclear antibody production is influenced by the presence of commensal gut flora, in particular increased colonization with segmented filamentous bacteria, and IL-17 receptor signaling. Together, these data indicate that neonatal colonization of gut microbiota influences generalized autoimmunity in adult life.


Subject(s)
Autoimmunity/immunology , Microbiota/immunology , Animals , Antibodies, Antinuclear/genetics , Antibodies, Antinuclear/immunology , Autoimmunity/genetics , Female , Flow Cytometry , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
7.
Biomacromolecules ; 15(6): 2301-9, 2014 Jun 09.
Article in English | MEDLINE | ID: mdl-24805802

ABSTRACT

During the past decade, extensive research has undeniably improved the formulation and delivery of oral vaccines. Nevertheless, several factors, such as the harsh gastrointestinal environment together with tolerance induction to exogenous antigens, have thus far impeded the optimal effectiveness and clinical application of oral delivery systems. The current study encompasses an initial evaluation of the stability, biocompatibility, and cellular uptake of two promising candidate systems for oral antigen delivery, that is, calcium carbonate- (CP) and mannitol-templated (MP) porous microspheres. Both spray-dried formulations were efficiently internalized by human intestinal epithelial cells (Caco-2 and HT-29) and degraded into phagolysosomal intracellular compartments. In addition, cellular particle uptake and processing significantly up-regulated the expression of (HLA) class-II and costimulatory molecules on intestinal epithelial cells. Even though the high surface-area-to-volume ratio of the microspheres was expected to favor protease access, antigen release was remarkably limited in simulated intestinal fluid and was even absent under gastric conditions. Finally, neither CP nor MP exerted cytotoxicity upon prolonged in vitro incubation with high antigen concentration. Altogether, these data support the potential of CP and MP for oral antigen delivery and motivate the further development of these promising carrier systems in in vivo studies.


Subject(s)
Antigens/metabolism , Biocompatible Materials/metabolism , Drug Delivery Systems/methods , Microspheres , Administration, Oral , Antigens/administration & dosage , Biocompatible Materials/administration & dosage , Caco-2 Cells , Cell Survival/drug effects , Cell Survival/physiology , Drug Stability , HT29 Cells , Humans , Ovalbumin/administration & dosage , Ovalbumin/metabolism , Serum Albumin, Bovine/administration & dosage , Serum Albumin, Bovine/metabolism
8.
Hum Vaccin Immunother ; 10(5): 1309-18, 2014.
Article in English | MEDLINE | ID: mdl-24553259

ABSTRACT

Oral vaccination is the most challenging vaccination method due to the administration route. However, oral vaccination has socio-economic benefits and provides the possibility of stimulating both humoral and cellular immune responses at systemic and mucosal sites. Despite the advantages of oral vaccination, only a limited number of oral vaccines are currently approved for human use. During the last decade, extensive research regarding antigen-based oral vaccination methods have improved immunogenicity and induced desired immunological outcomes. Nevertheless, several factors such as the harsh gastro-intestinal environment and oral tolerance impede the clinical application of oral delivery systems. To date, human clinical trials investigating the efficacy of these systems are still lacking. This review addresses the rationale and key biological and physicochemical aspects of oral vaccine design and highlights the use of yeast-derived ß-glucan microparticles as an oral vaccine delivery platform.


Subject(s)
Drug Discovery/trends , Saccharomyces cerevisiae/immunology , Vaccines/administration & dosage , Vaccines/immunology , beta-Glucans/administration & dosage , beta-Glucans/immunology , Administration, Oral , Animals , Drug Delivery Systems/trends , Humans , Microspheres , Yeasts/immunology
9.
J Control Release ; 172(3): 671-8, 2013 Dec 28.
Article in English | MEDLINE | ID: mdl-24041710

ABSTRACT

Continuously improving the developmental process and the efficacy of oral vaccines is essential in the fight against intestinal pathogens. A promising strategy for vaccination applying safe, biodegradable and non-replicating antigen delivery systems has gained increased interest for eliciting cellular and humoral immune responses. The current study evaluates the potential of ß-glucan particles (GP) as an oral antigen delivery system and their adjuvant characteristics. GP are efficiently internalized by human intestinal epithelial cell lines (Caco-2 and HT-29 cells), without exerting negative effects on cell viability. GP triggered the expression of pro-inflammatory cytokines IL-23p19, IL-8 and the ß-glucan receptors dectin-1 and TLR2 by activated Caco-2 cells, and CCL20 in HT-29 cells. In contrast, the expression level of TGF-ß, an important mediator of oral tolerance, was significantly downregulated in HT-29 cells. Additionally, adoptive transfer experiments showed proliferating ovalbumin (OVA)-specific CD4(+) T cells mainly in the spleens of GP-OVA-fed mice. Furthermore, we detected a significantly increased IL-17 and a trend towards increased IFN-γ production in the spleen of GP-OVA-fed mice upon antigen restimulation. Oral administration of GP-OVA induced increased OVA-specific IgA, secretory-IgA (S-IgA) and secretory component (SC) production in intestinal fluids. Our data show that GP vehicles are able to deliver OVA via an oral route allowing efficient antigen presentation alongside adaptive immune activation, resulting in a Th17-biased response and the production of OVA-specific IgA, secretory-IgA and secretory component antibodies.


Subject(s)
Antigens/administration & dosage , Ovalbumin/administration & dosage , Vaccines/administration & dosage , beta-Glucans/immunology , Administration, Oral , Animals , Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Caco-2 Cells , Cytokines/immunology , HT29 Cells , Humans , Immunoglobulin A/immunology , Male , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Vaccination , Vaccines/immunology , beta-Glucans/chemistry
10.
J Crohns Colitis ; 6(1): 1-12, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22261522

ABSTRACT

Epidemiological evidence demonstrates that smoking is the most important environmental risk factor in Crohn's disease while it positively interferes with the disease course of ulcerative colitis. However, the underlying mechanisms through which smoking exerts this divergent effect and affects pathogenesis of inflammatory bowel disease are largely unknown. Animal smoke models are good models to investigate the impact of cigarette smoke on intestinal physiology and inflammation. They enable one to explore the interaction of smoke components and the gut on cellular and molecular level, clarifying how smoking interferes with normal gut function and with disease course in inflammatory conditions. This review describes the currently used animal models for studying the impact of cigarette smoke on the intestinal tract. We first discuss the different methods for simulation of smoking. Furthermore, we focus on the effect of smoke exposure on normal gut physiology and immunology, on experimental (entero)colitis, and on inflammation-induced neoplasia. Based on this current knowledge, a hypothesis is formulated about the mechanisms through which cigarette smoke interferes with the gut in normal and pathological conditions.


Subject(s)
Inflammatory Bowel Diseases/physiopathology , Intestines/drug effects , Models, Animal , Nicotiana/adverse effects , Nicotine/adverse effects , Smoking/adverse effects , Animals , Humans , Inflammatory Bowel Diseases/immunology , Intestinal Neoplasms/physiopathology , Intestines/pathology , Intestines/physiopathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Risk Factors
11.
Histochem Cell Biol ; 137(3): 293-301, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22198275

ABSTRACT

Cigarette smoke (CS) exposure is associated with increased autophagy in several cell types, such as bronchial epithelial cells. Smoking is also an environmental risk factor in Crohn's disease, in which impairment of the autophagy-mediated anti-bacterial pathway has been implicated. So far, it is unknown whether CS induces autophagy in the gut. Here, we examined the effect of chronic CS exposure on autophagy in the follicle-associated epithelium (FAE) of murine Peyer's patches. Transmission electron microscopy revealed that the proportion of cell area occupied by autophagic vesicles significantly increased in the FAE after CS exposure. An increased number of autophagic vesicles was observed in the FAE, whereas the vesicle size remained unaltered. Besides enterocytes, also M-cells contain more autophagic vesicles upon CS exposure. In addition, the mRNA level of the autophagy-related protein Atg7 in the underlying Peyer's patches is increased after CS exposure, which indicates that the autophagy-inducing effect of CS is not limited to the FAE. In conclusion, our results demonstrate that CS exposure induces autophagy in murine FAE and in the underlying immune cells of Peyer's patches, suggesting that CS exposure increases the risk for Crohn's disease by causing epithelial oxidative damage, which needs to be repaired by autophagy.


Subject(s)
Autophagy/physiology , Ileum/pathology , Intestinal Mucosa/pathology , Peyer's Patches/pathology , Tobacco Smoke Pollution/adverse effects , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Beclin-1 , Chronic Disease , Crohn Disease/epidemiology , Crohn Disease/pathology , Ileum/ultrastructure , Intestinal Mucosa/ultrastructure , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Peyer's Patches/ultrastructure , RNA, Messenger/metabolism , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...