Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Pharmacol Rep ; 74(1): 111-123, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34757518

ABSTRACT

BACKGROUND: Obesity is still a worldwide public health problem, requiring the development of adjuvant therapies to combat it. In this context, modulation of the intestinal microbiota seems prominent, given that the composition of the intestinal microbiota contributes to the outcome of this disease. The aim of this work is to investigate the treatment with an antimicrobial and/or a potential probiotic against overweight. METHODS: Male C57BL/6J mice were subjected to a 12-week overweight induction protocol. After that, 4-week treatment was started, with mice divided into four groups: control, treated with distilled water; potential probiotic, with Lactobacillus gasseri LG-G12; antimicrobial, with ceftriaxone; and antimicrobial + potential probiotic with ceftriaxone in the first 2 weeks and L. gasseri LG-G12 in the subsequent weeks. RESULTS: The treatment with ceftriaxone in isolated form or in combination with the potential probiotic provided a reduction in body fat. However, such effect is supposed to be a consequence of the negative action of ceftriaxone on the intestinal microbiota composition, and this intestinal dysbiosis may have contributed to the destruction of the intestinal villi structure, which led to a reduction in the absorptive surface. Also, the effects of L. gasseri LG-G12 apparently have been masked by the consumption of the high-fat diet. CONCLUSIONS: The results indicate that the use of a ceftriaxone in the adjuvant treatment of overweight is not recommended due to the potential risk of developing inflammatory bowel disease.


Subject(s)
Ceftriaxone/pharmacology , Dysbiosis , Gastrointestinal Microbiome , Intestinal Absorption , Obesity , Adjuvants, Pharmaceutic/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Dysbiosis/chemically induced , Dysbiosis/immunology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Inflammatory Bowel Diseases/immunology , Intestinal Absorption/drug effects , Intestinal Absorption/immunology , Lactobacillus gasseri/physiology , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/microbiology , Probiotics/pharmacology , Risk Assessment
2.
Clin Transl Sci ; 14(2): 729-736, 2021 03.
Article in English | MEDLINE | ID: mdl-33278326

ABSTRACT

Changes in absorptive capacity and first-pass metabolism in the small intestine affect oral drug bioavailability. Characterization of such changes as a consequence of inflammation is important for developing physiologically-based pharmacokinetic (PBPK) models for inflammatory bowel disease. We sought to elucidate the impact of small intestinal Crohn's disease (CD) on villous length and CYP3A4 expression in children. Freshly frozen duodenal and terminal ileum (TI) biopsies from 107 children (1-19 years) with and without CD were evaluated for active inflammation. Villous length and CYP3A4 mRNA/protein expression were compared among regions of active and inactive inflammation in CD and controls. A twofold reduction in villous length was observed in inflamed duodena and ilia of children with CD, but in the absence of regional inflammation, villi in CD were comparable in length to controls. Expression of CYP3A4 mRNA correlated significantly with villous length in the TI (P = 0.0003), with a trend observed in the duodenum that did not reach statistical significance. In the presence of active inflammation, a significant decrease in CYP3A protein expression was confirmed in the duodenum, where protein expression also correlated significantly with villous length across diagnoses (P < 0.0001). Our findings suggest that previous observations of decreased CYP3A4 expression and function in inflamed intestine may not be due solely to downregulation by inflammatory cytokines, but also to villous blunting and subsequent loss of surface area for protein expression. This information is relevant for PBPK model development and could aid with dose adjustment decisions for oral CYP3A4 substrates administered during CD flare (e.g., budesonide).


Subject(s)
Anti-Inflammatory Agents/pharmacokinetics , Crohn Disease/drug therapy , Cytochrome P-450 CYP3A/metabolism , Intestinal Mucosa/metabolism , Administration, Oral , Adolescent , Anti-Inflammatory Agents/administration & dosage , Biological Availability , Biopsy , Budesonide/administration & dosage , Budesonide/pharmacokinetics , Case-Control Studies , Child , Child, Preschool , Crohn Disease/immunology , Crohn Disease/pathology , Dose-Response Relationship, Drug , Duodenum/cytology , Duodenum/immunology , Duodenum/metabolism , Duodenum/pathology , Female , Humans , Ileum/cytology , Ileum/immunology , Ileum/metabolism , Ileum/pathology , Infant , Intestinal Absorption/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Male , Models, Biological , Young Adult
3.
Curr Pharm Biotechnol ; 21(15): 1603-1615, 2020.
Article in English | MEDLINE | ID: mdl-32410561

ABSTRACT

In recent decades, there has been a very rapid increase in the prevalence of diabetes globally, with serious health and economic implications. Although today there are several therapeutic treatments for this disease, these do not address the causes of the disease and have serious side effects, so it is necessary to seek new treatments to replace or complement the existing ones. Among these complementary treatments, a strong link between the intestinal microbiota and diabetes has been demonstrated, which has focused attention on the use of biotherapy to regulate the function of the intestinal microbiota and, thus, treat diabetes. In this way, the main objective of this work is to provide a review of the latest scientific evidence on diabetes, gathering information about new trends in its management, and especially, the influence of the intestinal microbiota and microbiome on this pathology. It is possible to conclude that the relationship between the intestinal microbiota and diabetes is carried out through alterations in energy metabolism, the immune system, changes in intestinal permeability, and a state of low-intensity systemic inflammation. Although, currently, most of the experimental work, using probiotics for diabetes management, has been done on experimental animals, the results obtained are promising. Thus, the modification of the microbiota through biotherapy has shown to improve the symptoms and severity of diabetes through various mechanisms related to these alterations.


Subject(s)
Diabetes Mellitus/drug therapy , Gastrointestinal Microbiome/drug effects , Prebiotics/administration & dosage , Probiotics/therapeutic use , Animals , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Diabetes Mellitus/microbiology , Energy Metabolism/drug effects , Humans , Inflammation , Intestinal Absorption/drug effects , Intestinal Absorption/immunology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Permeability
4.
Nature ; 579(7800): 575-580, 2020 03.
Article in English | MEDLINE | ID: mdl-32050257

ABSTRACT

The intestinal mucosa serves both as a conduit for the uptake of food-derived nutrients and microbiome-derived metabolites, and as a barrier that prevents tissue invasion by microorganisms and tempers inflammatory responses to the myriad contents of the lumen. How the intestine coordinates physiological and immune responses to food consumption to optimize nutrient uptake while maintaining barrier functions remains unclear. Here we show in mice how a gut neuronal signal triggered by food intake is integrated with intestinal antimicrobial and metabolic responses that are controlled by type-3 innate lymphoid cells (ILC3)1-3. Food consumption rapidly activates a population of enteric neurons that express vasoactive intestinal peptide (VIP)4. Projections of VIP-producing neurons (VIPergic neurons) in the lamina propria are in close proximity to clusters of ILC3 that selectively express VIP receptor type 2 (VIPR2; also known as VPAC2). Production of interleukin (IL)-22 by ILC3, which is upregulated by the presence of commensal microorganisms such as segmented filamentous bacteria5-7, is inhibited upon engagement of VIPR2. As a consequence, levels of antimicrobial peptide derived from epithelial cells are reduced but the expression of lipid-binding proteins and transporters is increased8. During food consumption, the activation of VIPergic neurons thus enhances the growth of segmented filamentous bacteria associated with the epithelium, and increases lipid absorption. Our results reveal a feeding- and circadian-regulated dynamic neuroimmune circuit in the intestine that promotes a trade-off between innate immune protection mediated by IL-22 and the efficiency of nutrient absorption. Modulation of this pathway may therefore be effective for enhancing resistance to enteropathogens2,3,9 and for the treatment of metabolic diseases.


Subject(s)
Eating/physiology , Immunity, Innate/immunology , Intestinal Absorption/physiology , Intestines/immunology , Intestines/physiology , Lymphocytes/immunology , Neurons/metabolism , Vasoactive Intestinal Peptide/metabolism , Animals , Circadian Rhythm/physiology , Eating/immunology , Female , Interleukins/biosynthesis , Interleukins/immunology , Intestinal Absorption/immunology , Intestines/cytology , Intestines/microbiology , Lymphocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Postprandial Period/physiology , Receptors, CCR6/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Symbiosis , Interleukin-22
5.
Curr Protein Pept Sci ; 21(8): 777-784, 2020.
Article in English | MEDLINE | ID: mdl-31889482

ABSTRACT

Medium-chain fatty acids (MCFAs) are the main form of Medium Chain Triglycerides (MCTs) utilized by monogastric animals. MCFAs can be directly absorbed and supply rapid energy to promote the renewal and repair of intestinal epithelial cells, maintain the integrity of intestinal mucosal barrier function, and reduce inflammation and stress. In our review, we pay more attention to the role of MCFAs on intestinal microbiota and mucosa immunity to explore MCFA's positive effect. It was found that MCFAs and their esterified forms can decrease pathogens while increasing probiotics. In addition, being recognized via specific receptors, MCFAs are capable of alleviating inflammation to a certain extent by regulating inflammation and immune-related pathways. MCFAs may also have a certain value to relieve intestinal allergy and inflammatory bowel disease (IBD). Unknown mechanism of various MCFA characteristics still causes dilemmas in the application, thus MCFAs are used generally in limited dosages and combined with short-chain organic acids (SOAs) to attain ideal results. We hope that further studies will provide guidance for the practical use of MCFAs in animal feed.


Subject(s)
Caprylates/immunology , Colitis, Ulcerative/diet therapy , Crohn Disease/diet therapy , Decanoic Acids/immunology , Irritable Bowel Syndrome/diet therapy , Lauric Acids/immunology , Animal Feed/analysis , Animals , Caprylates/administration & dosage , Caprylates/metabolism , Colitis, Ulcerative/immunology , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Crohn Disease/immunology , Crohn Disease/microbiology , Crohn Disease/pathology , Cytokines/genetics , Cytokines/immunology , Decanoic Acids/administration & dosage , Decanoic Acids/metabolism , Gene Expression Regulation/drug effects , Humans , Immunity, Mucosal/drug effects , Intestinal Absorption/drug effects , Intestinal Absorption/immunology , Intestines/drug effects , Intestines/immunology , Intestines/microbiology , Irritable Bowel Syndrome/immunology , Irritable Bowel Syndrome/microbiology , Irritable Bowel Syndrome/pathology , Lauric Acids/administration & dosage , Lauric Acids/metabolism , NF-kappa B/genetics , NF-kappa B/immunology , Stomach/drug effects , Stomach/immunology , Stomach/microbiology , Triglycerides/immunology , Triglycerides/metabolism
6.
J Crohns Colitis ; 14(3): 316-322, 2020 Mar 13.
Article in English | MEDLINE | ID: mdl-31665264

ABSTRACT

BACKGROUND AND AIMS: Anaemia is common in patients with inflammatory bowel disease [IBD], its two main aetiologies being iron deficiency anaemia [IDA] and anaemia of chronic inflammation [ACI]. Impaired intestinal iron absorption due to inflammatory cytokines is thought to play a role in ACI. We undertook for the first time a controlled prospective study investigating effects of differing underlying diseases, disease locations, and types of iron deficiency or anaemia on oral iron absorption in adult IBD patients with and without inflammation. METHODS: This study was a comparative, single-centred open clinical trial in adults with IBD [n = 73] and healthy controls [n = 22]. Baseline parameters included blood count, iron status [ferritin, transferrin, transferrin saturation, soluble transferrin receptor, hepcidin, serum iron], high-sensitivity C-reactive protein [hsCRP] and interleukin-6. Iron absorption was tested using one oral, enteric-coated capsule containing 567.7 mg iron[II]-glycine-sulphate complex. Serum iron was determined 60/90/120/180/240 min after ingestion. RESULTS: Iron absorption capacity was shown to be influenced by inflammation and anaemia or iron deficiency [ID] type but not by underlying disease type or localisation. The ACI group showed a significantly lower iron absorption capacity than all others. Whereas hsCRP levels [-0.387, p < 0.001], IL-6 [-0.331, p = 0.006], ferritin [-0.531, p < 0.001], and serum hepcidin [-0.353, p = 0.003] correlated negatively with serum iron change at 2 h, transferrin showed a positive correlation at the same time point [0.379, p < 0.001]. CONCLUSIONS: Underlying disease type and localisation appear to have little effect on iron absorption capacity, whereas lack of response to oral iron correlates well with serum markers of inflammation. Iron absorption capacity is thus significantly reduced in the presence of inflammation.


Subject(s)
Anemia, Iron-Deficiency , Ferritins/blood , Inflammation/blood , Inflammatory Bowel Diseases , Interleukin-6/blood , Iron , Oral Mucosal Absorption/immunology , Transferrin/analysis , Adult , Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/etiology , Anemia, Iron-Deficiency/immunology , Biomarkers/blood , C-Reactive Protein/analysis , Correlation of Data , Female , Germany/epidemiology , Hepcidins/blood , Humans , Inflammatory Bowel Diseases/blood , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/epidemiology , Intestinal Absorption/immunology , Iron/blood , Iron Deficiencies , Male , Middle Aged
7.
Front Immunol ; 11: 599637, 2020.
Article in English | MEDLINE | ID: mdl-33542716

ABSTRACT

Food allergy is a potentially fatal disease affecting 8% of children and has become increasingly common in the past two decades. Despite the prevalence and severe nature of the disease, the mechanisms underlying sensitization remain to be further elucidated. The Collaborative Cross is a genetically diverse panel of inbred mice that were specifically developed to study the influence of genetics on complex diseases. Using this panel of mouse strains, we previously demonstrated CC027/GeniUnc mice, but not C3H/HeJ mice, develop peanut allergy after oral exposure to peanut in the absence of a Th2-skewing adjuvant. Here, we investigated factors associated with sensitization in CC027/GeniUnc mice following oral exposure to peanut, walnut, milk, or egg. CC027/GeniUnc mice mounted antigen-specific IgE responses to peanut, walnut and egg, but not milk, while C3H/HeJ mice were not sensitized to any antigen. Naïve CC027/GeniUnc mice had markedly lower total fecal IgA compared to C3H/HeJ, which was accompanied by stark differences in gut microbiome composition. Sensitized CC027/GeniUnc mice had significantly fewer CD3+ T cells but higher numbers of CXCR5+ B cells and T follicular helper cells in the mesenteric lymph nodes compared to C3H/HeJ mice, which is consistent with their relative immunoglobulin production. After oral challenge to the corresponding food, peanut- and walnut-sensitized CC027/GeniUnc mice experienced anaphylaxis, whereas mice exposed to milk and egg did not. Ara h 2 was detected in serum collected post-challenge from peanut-sensitized mice, indicating increased absorption of this allergen, while Bos d 5 and Gal d 2 were not detected in mice exposed to milk and egg, respectively. Machine learning on the change in gut microbiome composition as a result of food protein exposure identified a unique signature in CC027/GeniUnc mice that experienced anaphylaxis, including the depletion of Akkermansia. Overall, these results demonstrate several factors associated with enteral sensitization in CC027/GeniUnc mice, including diminished total fecal IgA, increased allergen absorption and altered gut microbiome composition. Furthermore, peanuts and tree nuts may have inherent properties distinct from milk and eggs that contribute to allergy.


Subject(s)
Allergens/immunology , Feces/microbiology , Gastrointestinal Microbiome/immunology , Immunoglobulin A/immunology , Intestinal Absorption/immunology , Peanut Hypersensitivity , Allergens/genetics , Animals , Gastrointestinal Microbiome/genetics , Genetic Predisposition to Disease , Immunoglobulin A/genetics , Intestinal Absorption/genetics , Mice , Mice, Transgenic , Peanut Hypersensitivity/genetics , Peanut Hypersensitivity/immunology , Peanut Hypersensitivity/microbiology
8.
Nat Commun ; 10(1): 4517, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31586069

ABSTRACT

Neonatal inflammatory diseases are associated with severe morbidity, but the inflammatory factors underlying them and their potential effector mechanisms are poorly defined. Here we show that necrotizing enterocolitis in neonate mice is accompanied by elevation of IL-23 and IL-22 and decreased production of pancreatic enzymes. These phenotypes are mirrored in neonate mice overexpressing IL-23 in CX3CR1+ myeloid cells or in keratinocytes. The mice fail to grow and die prematurely, displaying systemic inflammation, nutrient malabsorption and decreased expression of intestinal and pancreatic genes mediating digestion and absorption of carbohydrates, proteins, and lipids. Germ-free environment improves, and genetic ablation of IL-22 restores normal growth in mice overexpressing IL-23. Mechanistically, IL-22 acts directly at the level of pancreatic acinar cells to decrease expression of the pancreas associated transcription factor 1a (PTF1a). These results show that augmented production of IL-23 and IL-22 in early life has a negative impact on pancreatic enzyme secretion and food absorption.


Subject(s)
Enterocolitis, Necrotizing/immunology , Interleukin-23/metabolism , Interleukins/metabolism , Pancreas/enzymology , Transcription Factors/metabolism , Acinar Cells/enzymology , Animals , Animals, Newborn , Cells, Cultured , Disease Models, Animal , Enterocolitis, Necrotizing/pathology , Humans , Interleukin-23/genetics , Interleukin-23/immunology , Interleukins/genetics , Interleukins/immunology , Intestinal Absorption/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Keratinocytes , Mice , Mice, Knockout , Myeloid Cells , Pancreas/cytology , Primary Cell Culture , Interleukin-22
9.
Am J Gastroenterol ; 114(10): 1587-1592, 2019 10.
Article in English | MEDLINE | ID: mdl-31274511

ABSTRACT

Celiac disease is a common inflammatory disease triggered by dietary gluten in genetically susceptible individuals. The strongest and best-characterized genetic susceptibilities in celiac disease are class II human leukocyte antigen (HLA) genes known as HLA-DQ2 and DQ8. HLA genetic testing is available through a number of commercial and academic laboratories and is used in the evaluation of celiac disease and to identify at-risk family members. Importantly, HLA genetic testing has a high negative predictive value for celiac disease, but a low positive predictive value. Therefore, for a practicing clinician, it is important to understand when to order HLA genetic testing, what test to order, and how to interpret the result. This review provides a practical primer on HLA genetics in celiac disease.


Subject(s)
Celiac Disease/diagnosis , Genetic Testing/standards , HLA-DQ Antigens/genetics , Practice Guidelines as Topic , Biomarkers/blood , Biopsy , Celiac Disease/blood , Celiac Disease/genetics , Celiac Disease/immunology , Duodenum/immunology , Duodenum/metabolism , Duodenum/pathology , Gastroenterology/standards , Genetic Predisposition to Disease , Glutens/immunology , Glutens/metabolism , HLA-DQ Antigens/immunology , Humans , Intestinal Absorption/genetics , Intestinal Absorption/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Predictive Value of Tests
10.
Clin Transl Gastroenterol ; 10(5): 1-10, 2019 05 22.
Article in English | MEDLINE | ID: mdl-31082858

ABSTRACT

OBJECTIVES: Celiac disease (CD) is a chronic enteropathy characterized by an autoimmune reaction in the small intestine of genetically susceptible individuals. The underlying causes of autoimmune reaction and its effect on host metabolism remain largely unknown. Herein, we apply lipidomics to elucidate the early events preceding clinical CD in a cohort of Finnish children, followed up in the Type 1 Diabetes Prediction and Prevention study. METHODS: Mass spectrometry-based lipidomics profiling was applied to a longitudinal/prospective series of 233 plasma samples obtained from CD progressors (n = 23) and healthy controls (n = 23), matched for human leukocyte antigen (HLA) risk, sex, and age. The children were followed from birth until diagnosis of clinical CD and subsequent introduction of a gluten-free diet. RESULTS: Twenty-three children progressed to CD at a mean age of 4.8 years. They showed increased amounts of triacylglycerols (TGs) of low carbon number and double bond count and a decreased level of phosphatidylcholines by age 3 months as compared to controls. These differences were exacerbated with age but were not observed at birth (cord blood). No significant differences were observed in the essential TGs. DISCUSSION: Our preliminary findings suggest that abnormal lipid metabolism associates with the development of clinical CD and occurs already before the first introduction of gluten to the diet. Moreover, our data suggest that the specific TGs found elevated in CD progressors may be due to a host response to compromised intake of essential lipids in the small intestine, requiring de novo lipogenesis.


Subject(s)
Celiac Disease/diagnosis , Glutens/immunology , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Triglycerides/blood , Autoimmunity , Biopsy , Celiac Disease/blood , Celiac Disease/diet therapy , Celiac Disease/immunology , Child, Preschool , Diet, Gluten-Free , Disease Progression , Endoscopy, Gastrointestinal , Finland , Glutens/administration & dosage , HLA-DQ Antigens/blood , HLA-DQ Antigens/immunology , Humans , Infant , Intestinal Absorption/immunology , Intestinal Mucosa/diagnostic imaging , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestine, Small/diagnostic imaging , Intestine, Small/immunology , Intestine, Small/pathology , Lipid Metabolism/immunology , Lipidomics , Longitudinal Studies , Prospective Studies , Triglycerides/metabolism
11.
Am J Physiol Cell Physiol ; 312(4): C438-C445, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28249988

ABSTRACT

Association between circulating lipopolysaccharide (LPS) and metabolic diseases (such as type 2 diabetes and atherosclerosis) has shifted the focus from high-fat high-cholesterol containing Western-type diet (WD)-induced changes in gut microbiota per se to release of gut bacteria-derived products (e.g., LPS) into circulation due to intestinal barrier dysfunction as the possible mechanism for the chronic inflammatory state underlying the development of these diseases. We demonstrated earlier that oral supplementation with curcumin attenuates WD-induced development of type 2 diabetes and atherosclerosis. Poor bioavailability of curcumin has precluded the establishment of a causal relationship between oral supplementation and it is in vivo effects. We hypothesized that curcumin attenuates WD-induced chronic inflammation and associated metabolic diseases by modulating the function of intestinal epithelial cells (IECs) and the intestinal barrier function. The objective of the present study was to delineate the underlying mechanisms. The human IEC lines Caco-2 and HT-29 were used for these studies and modulation of direct as well as indirect effects of LPS on intracellular signaling as well as tight junctions were examined. Pretreatment with curcumin significantly attenuated LPS-induced secretion of master cytokine IL-1ß from IECs and macrophages. Furthermore, curcumin also reduced IL-1ß-induced activation of p38 MAPK in IECs and subsequent increase in expression of myosin light chain kinase involved in the phosphorylation of tight junction proteins and ensuing disruption of their normal arrangement. The major site of action of curcumin is, therefore, likely the IECs and the intestinal barrier, and by reducing intestinal barrier dysfunction, curcumin modulates chronic inflammatory diseases despite poor bioavailability.


Subject(s)
Cell Communication/immunology , Colitis/immunology , Curcumin/administration & dosage , Intestinal Absorption/immunology , Intestinal Mucosa/immunology , Tight Junctions/immunology , Caco-2 Cells , Cell Communication/drug effects , Colitis/chemically induced , Colitis/drug therapy , Colon/drug effects , Colon/immunology , Dose-Response Relationship, Drug , Epithelial Cells , HT29 Cells , Humans , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Lipopolysaccharides , Tight Junctions/drug effects
12.
Gastroenterology ; 151(5): 923-932, 2016 11.
Article in English | MEDLINE | ID: mdl-27436071

ABSTRACT

BACKGROUND & AIMS: The gut microbiota affects intestinal permeability and mucosal mast cells (MMCs) responses. Activation of MMCs has been associated with absorption of dietary fat. We investigated whether the gut microbiota contributes to the fat-induced activation of MMCs in rats, and how antibiotics might affect this process. METHODS: Adult male Sprague-Dawley rats were given streptomycin and penicillin for 4 days (n = 6-8) to reduce the abundance of their gut flora, or normal drinking water (controls, n = 6-8). They underwent lymph fistula surgery and after an overnight recovery were given an intraduodenal bolus of intralipid. We collected intestinal tissues and lymph fluid and assessed activation of MMCs, intestinal permeability, and fat transport parameters. RESULTS: Compared with controls, intestinal lymph from rats given antibiotics had reduced levels of mucosal mast cell protease II (produced by MMCs) and decreased activity of diamine oxidase (produced by enterocytes) (P < .05). Rats given antibiotics had reduced intestinal permeability in response to dietary lipid compared with controls (P < .01). Unexpectedly, antibiotics also reduced lymphatic transport of triacylglycerol and phospholipid (P < .01), concomitant with decreased levels of mucosal apolipoproteins B, A-I, and A-IV (P < .01). No differences were found in intestinal motility or luminal pancreatic lipase activity between rats given antibiotics and controls. These effects were not seen with an acute dose of antibiotics or 4 weeks after the antibiotic regimen ended. CONCLUSIONS: The intestinal microbiota appears to activate MMCs after the ingestion of fat in rats; this contributes to fat-induced intestinal permeability. We found that the gut microbiome promotes absorption of lipid, probably by intestinal production of apolipoproteins and secretion of chylomicrons.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dietary Fats/metabolism , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Mast Cells/drug effects , Penicillins/pharmacology , Streptomycin/pharmacology , Animals , Anti-Bacterial Agents/administration & dosage , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Intestinal Absorption/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Mast Cells/metabolism , Mast Cells/microbiology , Penicillins/administration & dosage , Permeability , Rats , Rats, Sprague-Dawley , Streptomycin/administration & dosage
13.
Lipids Health Dis ; 15: 108, 2016 Jun 18.
Article in English | MEDLINE | ID: mdl-27317359

ABSTRACT

Obesity and its associated complications like type 2 diabetes (T2D) are reaching epidemic stages. Increased food intake and lack of exercise are two main contributing factors. Recent work has been highlighting an increasingly more important role of gut microbiota in metabolic disorders. It's well known that gut microbiota plays a major role in the development of food absorption and low grade inflammation, two key processes in obesity and diabetes. This review summarizes key discoveries during the past decade that established the role of gut microbiota in the development of obesity and diabetes. It will look at the role of key metabolites mainly the short chain fatty acids (SCFA) that are produced by gut microbiota and how they impact key metabolic pathways such as insulin signalling, incretin production as well as inflammation. It will further look at the possible ways to harness the beneficial aspects of the gut microbiota to combat these metabolic disorders and reduce their impact.


Subject(s)
Diabetes Mellitus, Type 2/microbiology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/microbiology , Lipid Metabolism/immunology , Obesity/microbiology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/pathology , Fatty Acids, Volatile/biosynthesis , Fatty Acids, Volatile/metabolism , Gastrointestinal Tract/immunology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Gene Expression Regulation , Humans , Incretins/genetics , Incretins/metabolism , Inflammation , Insulin/genetics , Insulin/metabolism , Intestinal Absorption/immunology , Obesity/complications , Obesity/immunology , Obesity/pathology , Signal Transduction
14.
J Hepatobiliary Pancreat Sci ; 23(11): 681-687, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27168084

ABSTRACT

BACKGROUND: The aim of the present study was to investigate the role of early enteral nutrition (EEN) in the intestinal immune barrier in severe acute pancreatitis (SAP), and to explore its potential mechanisms. METHODS: Sixty rats were randomly assigned to three groups: sham-operated group (SO group, n = 20), SAP group receiving EEN (SAP + EEN group, n = 20), and SAP group receiving total parental nutrition (SAP + TPN group, n = 20). SAP was induced by infusion of sodium taurocholate. Rats were killed 5 days after nutritional support. The pathological damage of the intestine was determined using HE staining. The expression of MAdCAM-1, CD4+ , and CD8+ in Peyer's lymph nodes of the distal ilium was examined by immunohistochemistry. Serum levels of endotoxin and bacterial translocation were determined. RESULTS: The survival rate in the SAP + TPN (50%) and SAP + EEN (75%) groups was significantly lower than in the SO group (100%) (P < 0.05). The survival rate in the SAP + EEN group was significantly higher than in the SAP + TPN group (P < 0.05). The expression of MAdCAM-1, CD4+ and CD8+ in the intestine was decreased in SAP rats. EEN significantly increased the expression of MAdCAM-1, CD4+ and CD8+ compared with TPN, accompanied by a decrease in the serum levels of endotoxin and bacterial translocation. CONCLUSIONS: Early enteral nutrition improves intestinal immune barrier, thus reducing bacterial and endotoxin translocation and improving the survival rate in SAP rats.


Subject(s)
Enteral Nutrition/methods , Intestinal Absorption/immunology , Intestinal Mucosa/metabolism , Pancreatitis, Acute Necrotizing/therapy , Amylases/blood , Analysis of Variance , Animals , Biopsy, Needle , Disease Models, Animal , Endotoxins/metabolism , Female , Ilium/pathology , Immunohistochemistry , Intestinal Mucosa/pathology , Pancreatitis, Acute Necrotizing/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Reference Values , Severity of Illness Index , Survival Rate
16.
Biochem Biophys Res Commun ; 473(4): 1090-1093, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27060548

ABSTRACT

A lectin like protein designated as LSMT is recently discovered in Agaricus bisporus. The protein adopts very similar structure to Ricin-B like lectin from Clitocybe nebularis (CNL) and HA-33 from Clostridium botulinum (HA-33), which both recognize sugar molecules that decorate the surface of the epithelial cells of the intestine. A preliminary study in silico pointed out potential capability of LSMT to perform such biological activity. Following that hypothesis, we demonstrated that LSMT is indeed capable of penetrating out from a dialysis tube of the mice intestine origin. Furthermore, the protein appeared not to evoke the immune response upon introduction into mice, unlike its structural homologs. This is the first report on the biological implication of LSMT that might lead to its application.


Subject(s)
Immune Tolerance/immunology , Intestinal Absorption/immunology , Lectins/chemistry , Lectins/immunology , Models, Immunological , Animals , Computer Simulation , Female , Lectins/classification , Mice , Models, Chemical , Permeability , Sequence Analysis, Protein , Sequence Homology, Amino Acid
17.
J Microbiol Immunol Infect ; 48(6): 583-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25087197

ABSTRACT

BACKGROUND/PURPOSE: Whether absorption of verotoxin (VT) 2 from the intestine in mice is inhibited by administration bovine immune colostral antibody against VT2 was investigated. METHODS: Three-week-old mice were administered VT2 solution at 477.8 ng/mL or 955.6 ng/mL, and bovine immune colostral antibody against VT2 was then administered three times. Whey without antibody against VT2 was administered to control mice. Serum levels of VT2 were measured by fluorescence enzyme immunoassay. RESULTS: Serum levels of VT2 in mice administered VT2 solution at 477.8 ng/mL and bovine immune colostral antibody against VT2 scarcely changed. By contrast, serum levels of VT2 in control mice increased and peaked 12 hours after administration. Peak values were 15.4 ± 5.04 ng/mL. Furthermore, serum levels of VT2 at 12 hours and 16 hours in control mice were significantly higher than in mice administered bovine colostral antibody against VT2. Serum levels of VT2 in mice administered antibody at 955.6 ng/mL showed no significant differences between repeated administration of bovine immune colostral antibody and controls. CONCLUSION: These results suggest that absorption of VT2 from the intestine was inhibited by repeated administration of bovine immune colostral antibody against VT2 at early stages of Escherichia coli O157:H7 infection, whereas VT2 in the intestine remained at low levels.


Subject(s)
Colostrum/immunology , Immunoglobulin A, Secretory/immunology , Intestinal Absorption/immunology , Intestinal Mucosa/metabolism , Shiga Toxin 2/blood , Shiga Toxin 2/toxicity , Animals , Escherichia coli Infections/immunology , Escherichia coli Infections/prevention & control , Escherichia coli O157/immunology , Escherichia coli O157/pathogenicity , Foodborne Diseases/microbiology , Foodborne Diseases/prevention & control , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/pathology , Gerbillinae , Immunoglobulin A, Secretory/administration & dosage , Intestinal Absorption/physiology , Male , Mice , Mice, Inbred ICR , Shiga Toxin 2/metabolism
18.
Curr Oncol Rep ; 16(10): 406, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25123079

ABSTRACT

Humans depend on our commensal bacteria for nutritive, immune-modulating, and metabolic contributions to maintenance of health. However, this commensal community exists in careful balance that, if disrupted, enters dysbiosis; this has been shown to contribute to the pathogenesis of colon, gastric, esophageal, pancreatic, laryngeal, breast, and gallbladder carcinomas. This development is closely tied to host inflammation, which causes and is aggravated by microbial dysbiosis and increases vulnerability to pathogens. Advances in sequencing technology have increased our ability to catalog microbial species associated with various cancer types across the body. However, defining microbial biomarkers as cancer predictors presents multiple challenges, and existing studies identifying cancer-associated bacteria have reported inconsistent outcomes. Combining metabolites and microbiome analyses can help elucidate interactions between gut microbiota, metabolism, and the host. Ultimately, understanding how gut dysbiosis impacts host response and inflammation will be critical to creating an accurate picture of the role of the microbiome in cancer.


Subject(s)
Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Dysbiosis/pathology , Gram-Negative Bacterial Infections/pathology , Adaptive Immunity/immunology , Bacterial Load , Colorectal Neoplasms/immunology , Disease Progression , Dysbiosis/immunology , Dysbiosis/microbiology , Gene Expression Regulation, Bacterial , Gram-Negative Bacterial Infections/immunology , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Intestinal Absorption/immunology , Microbiota/immunology , Neoplasm Staging
19.
Internist (Berl) ; 55(8): 883-8, 2014 Aug.
Article in German | MEDLINE | ID: mdl-24831679

ABSTRACT

The challenge of the mucosal immune system is to develop tolerance toward intestinal antigens. Considering the quantity of bacteria that continuously attack the intestinal barrier, one can only imagine the complexity involved. To master this task, a tight network between the intestinal microbiota, the barrier, immune cells of the lamina propria as well as the adjacent mesenteric fat tissue is required. The key pathways involved have been revealed by the genome-wide association studies as well as functional data from experimental models. However, although knowledge with regard to the pathogenesis of chronic inflammatory bowel disease has been increasing continuously over recent decades, the current therapeutic strategies are limited to controlling the pro-inflammatory effector phase rather than achieving cure. The best example is cytokine neutralizing antibodies. The present review aims to describe the role of the various cell populations within the intestinal wall for disease pathogenesis and, thus, identify possible therapeutic strategies.


Subject(s)
Cytokines/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestines/immunology , Microbiota/immunology , Chronic Disease , Humans , Intestinal Absorption/immunology , Models, Immunological
20.
Inflamm Bowel Dis ; 20(6): 1049-56, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24788221

ABSTRACT

BACKGROUND: First-degree relatives of patients with Crohn's disease (CD) are at risk of developing the disease with 5% to 15% reported to be affected over time. Yet, a much greater proportion of them (>40%) shows features of "subclinical inflammation" with elevated intestinal inflammatory markers such as fecal calprotectin. The meaning of these findings is unclear in the absence of tissue data. METHODS: Thirty-eight asymptomatic first-degree relatives of patients with CD underwent ileocolonoscopy and other tests including fecal calprotectin. All known causes of intestinal inflammation were carefully excluded. Age and gender-matched controls consisted of 10 individuals who underwent colonoscopy for other reasons. Histology was scored based on known methods. RESULTS: Compared with controls, the relatives had significantly greater median values for fecal calprotectin and histological scores. In relatives, endoscopy identified 3 different phenotypes: (1) normal, (2) with minor lesions (aphthae or small superficial erosions), and (3) with typical CD inflammation. Based on the histological scores, the clustering analysis produced 3 corresponding highly separated clusters (61%, 26%, and 13% of the total, respectively) with divisive coefficient D = 0.94. When followed up (on the average for 53 mo), individuals in the second cluster had histological scores similar to baseline values (P = 0.12). CONCLUSIONS: Tissue studies in first-degree relatives of patients with CD reveal 3 distinct groups: normal, with minimal inflammation, and with frank disease. The second cluster represents a novel phenotype, which does not seem to develop the disease over time. These findings explain previous observations of "subclinical inflammation" in such population.


Subject(s)
Crohn Disease/epidemiology , Crohn Disease/genetics , Adult , Biomarkers/metabolism , Colon/immunology , Colon/pathology , Colonoscopy , Crohn Disease/pathology , Endoscopy, Gastrointestinal , Family , Feces/chemistry , Female , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Genetic Testing , Humans , Ileum/immunology , Ileum/pathology , Intestinal Absorption/immunology , Leukocyte L1 Antigen Complex/analysis , Male , Middle Aged , Phenotype , Risk Factors , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL