Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Rep ; 12(1): 6883, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35477719

ABSTRACT

Bone marrow transplantation (BMT) involves conditioning regimens which acutely induce side effects, including systemic inflammation, intestinal damage and shifts in the gut microbial composition, some of which may persist chronically. As the gut microbiota affect systemic immune responses, we aimed to investigate whether, post-BMT, the peripheral immune system is modulated as a direct consequence of alterations in the gut microbiota. We show that 24 weeks post-BMT, splenocytes but not peritoneal macrophages display increased cytokine response patterns upon ex-vivo stimulation with various pathogens as compared to untreated controls. The pattern of BMT-induced cytokine responses was transferred to splenocytes, and not to peritoneal macrophages, of healthy controls via co-housing and transferred to germfree mice via transplantation of cecum content. Thus, BMT induces changes in gut microbiota that in their turn increase cytokine responsiveness of splenocytes. Thus, BMT establishes a dominant microbiota that attenuates normalization of the immune-response.


Subject(s)
Gastrointestinal Microbiome , Animals , Bone Marrow Transplantation/adverse effects , Cytokines , Immune System , Mice , Spleen
2.
Mol Nutr Food Res ; 64(15): e1900732, 2020 08.
Article in English | MEDLINE | ID: mdl-31389129

ABSTRACT

SCOPE: Akkermansia muciniphila (A. muciniphila) is an intestinal commensal with anti-inflammatory properties both in the intestine and other organs. The aim is to investigate the effects of oral administration of A. muciniphila on lipid metabolism, immunity, and cuff-induced neointima formation in hyperlipidemic APOE*3-Leiden (E3L).CETP mice. METHODS AND RESULTS: Hyperlipidemic male E3L.CETP mice are daily treated with 2 × 108 CFU A. muciniphila by oral gavage for 4 weeks and the effects are determined on plasma lipid levels, immune parameters, and cuff-induced neointima formation and composition. A. muciniphila administration lowers body weight and plasma total cholesterol and triglycerides levels. A. muciniphila influences the immune cell composition in mesenteric lymph nodes, as evident from an increased total B cell population, while reducing the total T cell and neutrophil populations. Importantly, A. muciniphila reduces the expression of the activation markers MHCII on dendritic cells and CD86 on B cells. A. muciniphila also increases whole blood ex vivo lipopolysaccharide-stimulated IL-10 release. Finally, although treatment with A. muciniphila improves lipid metabolism and immunity, it does not affect neointima formation or composition. CONCLUSIONS: Four weeks of treatment with A. muciniphila exerts lipid-lowering and immunomodulatory effects, which are insufficient to inhibit neointima formation in hyperlipidemic E3L.CETP mice.


Subject(s)
Hyperlipidemias/therapy , Immunologic Factors/pharmacology , Lipids/blood , Probiotics/administration & dosage , Administration, Oral , Akkermansia/immunology , Akkermansia/physiology , Animals , Apolipoprotein E3/genetics , Disease Models, Animal , Hypercholesterolemia/immunology , Hypercholesterolemia/metabolism , Hypercholesterolemia/therapy , Hyperlipidemias/immunology , Hyperlipidemias/metabolism , Lipid Metabolism , Lipopolysaccharides/blood , Lymph Nodes/immunology , Male , Mice, Mutant Strains , Neointima/etiology , Neointima/prevention & control
3.
Sci Rep ; 8(1): 16515, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30409998

ABSTRACT

Gut microbiota have been implicated in the development of atherosclerosis and cardiovascular disease. Since the prebiotic inulin is thought to beneficially affect gut microbiota, we aimed to determine the effect of inulin supplementation on atherosclerosis development in APOE*3-Leiden.CETP (E3L.CETP) mice. Female E3L.CETP mice were fed a western-type diet containing 0.1% or 0.5% cholesterol with or without 10% inulin. The effects of inulin were determined on: microbiota composition, cecal short-chain fatty acid (SCFA) levels, plasma lipid levels, atherosclerosis development, hepatic morphology and hepatic inflammation. Inulin with 0.5% dietary cholesterol increased specific bacterial genera and elevated levels of cecal SCFAs, but did not affect plasma cholesterol levels or atherosclerosis development. Surprisingly, inulin resulted in mild hepatic inflammation as shown by increased expression of inflammation markers. However, these effects were not accompanied by increased hepatic macrophage number. Analogously, inulin induced mild steatosis and increased hepatocyte size, but did not affect hepatic triglyceride content. Inulin with 0.1% dietary cholesterol did not affect hepatic morphology, nor hepatic expression of inflammation markers. Overall, inulin did not reduce hypercholesterolemia or atherosclerosis development in E3L.CETP mice despite showing clear prebiotic activity, but resulted in manifestations of hepatic inflammation when combined with a high percentage of dietary cholesterol.


Subject(s)
Apolipoprotein E3/genetics , Atherosclerosis/immunology , Bacteria/drug effects , Gastrointestinal Microbiome/drug effects , Hypercholesterolemia/immunology , Inulin/administration & dosage , Prebiotics/administration & dosage , Animals , Apolipoprotein E3/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , Diet, Western/adverse effects , Disease Models, Animal , Fatty Acids/chemistry , Female , Hypercholesterolemia/chemically induced , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Inulin/pharmacology , Lipids/blood , Mice , Mice, Transgenic , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
4.
PLoS One ; 13(5): e0196165, 2018.
Article in English | MEDLINE | ID: mdl-29723205

ABSTRACT

The indigestible mannan oligosaccharides (MOS) derived from the outer cell wall of yeast Saccharomyces cerevisiae have shown potential to reduce inflammation. Since inflammation is one of the underlying mechanisms involved in the development of obesity-associated metabolic dysfunctions, we aimed to determine the effect of dietary supplementation with MOS on inflammation and metabolic homeostasis in lean and diet-induced obese mice. Male C57BL/6 mice were fed either a low fat diet (LFD) or a high fat diet (HFD) with, respectively, 10% or 45% energy derived from lard fat, with or without 1% MOS for 17 weeks. Body weight and composition were measured throughout the study. After 12 weeks of intervention, whole-body glucose tolerance was assessed and in week 17 immune cell composition was determined in mesenteric white adipose tissue (mWAT) and liver by flow cytometry and RT-qPCR. In LFD-fed mice, MOS supplementation induced a significant increase in the abundance of macrophages and eosinophils in mWAT. A similar trend was observed in hepatic macrophages. Although HFD feeding induced a classical shift from the anti-inflammatory M2-like macrophages towards the pro-inflammatory M1-like macrophages in both mWAT and liver from control mice, MOS supplementation had no effect on this obesity-driven immune response. Finally, MOS supplementation did not improve whole-body glucose homeostasis in both lean and obese mice.Altogether, our data showed that MOS had extra-intestinal immune modulatory properties in mWAT and liver. However these effects were not substantial enough to significantly ameliorate HFD-induced glucose intolerance or inflammation.


Subject(s)
Diet, High-Fat/adverse effects , Glucose Intolerance/immunology , Mannans/chemistry , Obesity/immunology , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Saccharomyces cerevisiae/chemistry , Adipose Tissue, White/drug effects , Adipose Tissue, White/immunology , Animals , Cell Count , Dietary Supplements , Eosinophils/cytology , Eosinophils/drug effects , Glucose Intolerance/chemically induced , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Liver/drug effects , Liver/immunology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/drug effects , Obesity/chemically induced
5.
Mol Nutr Food Res ; 62(10): e1700942, 2018 05.
Article in English | MEDLINE | ID: mdl-29665623

ABSTRACT

SCOPE: Mannan oligosaccharides (MOS) have proven effective at improving growth performance, while also reducing hyperlipidemia and inflammation. As atherosclerosis is accelerated both by hyperlipidemia and inflammation, we aim to determine the effect of dietary MOS on atherosclerosis development in hyperlipidemic ApoE*3-Leiden.CETP (E3L.CETP) mice, a well-established model for human-like lipoprotein metabolism. METHODS AND RESULTS: Female E3L.CETP mice were fed a high-cholesterol diet, with or without 1% MOS for 14 weeks. MOS substantially decreased atherosclerotic lesions up to 54%, as assessed in the valve area of the aortic root. In blood, IL-1RA, monocyte subtypes, lipids, and bile acids (BAs) were not affected by MOS. Gut microbiota composition was determined using 16S rRNA gene sequencing and MOS increased the abundance of cecal Bacteroides ovatus. MOS did not affect fecal excretion of cholesterol, but increased fecal BAs as well as butyrate in cecum as determined by gas chromatography mass spectrometry. CONCLUSION: MOS decreased the onset of atherosclerosis development via lowering of plasma cholesterol levels. These effects were accompanied by increased cecal butyrate and fecal excretion of BAs, presumably mediated via interactions of MOS with the gut microbiota.


Subject(s)
Atherosclerosis/diet therapy , Bile Acids and Salts/metabolism , Cholesterol/blood , Gastrointestinal Microbiome/drug effects , Mannans/pharmacology , Animals , Atherosclerosis/pathology , Bacteroides/isolation & purification , Biomarkers/metabolism , Butyrates/metabolism , Cecum/drug effects , Cecum/microbiology , Cholesterol/metabolism , Dietary Supplements , Feces , Female , Gastrointestinal Microbiome/physiology , Inflammation/diet therapy , Inflammation/metabolism , Liver/drug effects , Liver/metabolism , Mice, Mutant Strains , Triglycerides/blood
6.
Nutrients ; 10(2)2018 Feb 03.
Article in English | MEDLINE | ID: mdl-29401645

ABSTRACT

The prebiotic inulin has proven effective at lowering inflammation and plasma lipid levels. As atherosclerosis is provoked by both inflammation and hyperlipidemia, we aimed to determine the effect of inulin supplementation on atherosclerosis development in hypercholesterolemic APOE*3-Leiden (E3L) mice. Male E3L mice were fed a high-cholesterol (1%) diet, supplemented with or without 10% inulin for 5 weeks. At week 3, a non-constrictive cuff was placed around the right femoral artery to induce accelerated atherosclerosis. At week 5, vascular pathology was determined by lesion thickness, vascular remodeling, and lesion composition. Throughout the study, plasma lipids were measured and in week 5, blood monocyte subtypes were determined using flow cytometry analysis. In contrast to our hypothesis, inulin exacerbated atherosclerosis development, characterized by increased lesion formation and outward vascular remodeling. The lesions showed increased number of macrophages, smooth muscle cells, and collagen content. No effects on blood monocyte composition were found. Inulin significantly increased plasma total cholesterol levels and total cholesterol exposure. In conclusion, inulin aggravated accelerated atherosclerosis development in hypercholesterolemic E3L mice, accompanied by adverse lesion composition and outward remodeling. This process was not accompanied by differences in blood monocyte composition, suggesting that the aggravated atherosclerosis development was driven by increased plasma cholesterol.


Subject(s)
Apolipoprotein E3/metabolism , Atherosclerosis/pathology , Hypercholesterolemia/complications , Inulin/adverse effects , Prebiotics , Animals , Apolipoprotein E3/genetics , Atherosclerosis/genetics , Cholesterol/blood , Hypercholesterolemia/genetics , Ligation , Mice , Mice, Transgenic , Monocytes
7.
Methods Mol Biol ; 1730: 247-256, 2018.
Article in English | MEDLINE | ID: mdl-29363078

ABSTRACT

Short-chain fatty acids, the end products of fermentation of dietary fibers by the gut microbiota, have been shown to exert multiple effects on mammalian metabolism. For the analysis of short-chain fatty acids, gas chromatography-mass spectrometry is a very powerful and reliable method. Here, we describe a fast, reliable, and reproducible method for the separation and quantification of short-chain fatty acids in mouse feces, cecum content, and blood samples (i.e., plasma or serum) using gas chromatography-mass spectrometry. The short-chain fatty acids analyzed include acetic acid, propionic acid, butyric acid, valeric acid, hexanoic acid, and heptanoic acid.


Subject(s)
Cecum/chemistry , Fatty Acids, Volatile/analysis , Feces/chemistry , Metabolomics/methods , Acetic Acid/analysis , Acetic Acid/blood , Animals , Butyric Acid/analysis , Butyric Acid/blood , Caproates/analysis , Caproates/blood , Fatty Acids, Volatile/blood , Gas Chromatography-Mass Spectrometry , Heptanoic Acids/analysis , Heptanoic Acids/blood , Mice , Pentanoic Acids/analysis , Pentanoic Acids/blood , Propionates/analysis , Propionates/blood , Reproducibility of Results
8.
Methods Mol Biol ; 1730: 257-265, 2018.
Article in English | MEDLINE | ID: mdl-29363079

ABSTRACT

Our body contains a wide variety of fatty acids that differ in chain length, the degree of unsaturation, and location of the double bonds. As the various fatty acids play distinct roles in health and disease, methods that can specifically determine the fatty acid profile are needed for fundamental and clinical studies. Here we describe a method for the separation and quantification of fatty acids ranging from 8 to 24 carbon chain lengths in blood samples using gas chromatography-mass spectrometry following derivatization using pentafluorobenzyl bromide. This method quantitatively monitors fatty acid composition in a manner that satisfies the requirements for comprehensiveness, sensitivity, and accuracy.


Subject(s)
Fatty Acids/blood , Metabolomics/methods , Fluorobenzenes/chemistry , Gas Chromatography-Mass Spectrometry , Humans , Limit of Detection
9.
PLoS One ; 12(4): e0175524, 2017.
Article in English | MEDLINE | ID: mdl-28445487

ABSTRACT

Experimental bone marrow transplantation (BMT) in mice is commonly used to assess the role of immune cell-specific genes in various pathophysiological settings. The application of BMT in obesity research is hampered by the significant reduction in high-fat diet (HFD)-induced obesity. We set out to characterize metabolic tissues that may be affected by the BMT procedure and impair the HFD-induced response. Male C57BL/6 mice underwent syngeneic BMT using lethal irradiation. After a recovery period of 8 weeks they were fed a low-fat diet (LFD) or HFD for 16 weeks. HFD-induced obesity was reduced in mice after BMT as compared to HFD-fed control mice, characterized by both a reduced fat (-33%; p<0.01) and lean (-11%; p<0.01) mass, while food intake and energy expenditure were unaffected. As compared to control mice, BMT-treated mice had a reduced mature adipocyte volume (approx. -45%; p<0.05) and reduced numbers of preadipocytes (-38%; p<0.05) and macrophages (-62%; p<0.05) in subcutaneous, gonadal and visceral white adipose tissue. In BMT-treated mice, pancreas weight (-46%; p<0.01) was disproportionally decreased. This was associated with reduced plasma insulin (-68%; p<0.05) and C-peptide (-37%; p<0.01) levels and a delayed glucose clearance in BMT-treated mice on HFD as compared to control mice. In conclusion, the reduction in HFD-induced obesity after BMT in mice is at least partly due to alterations in the adipose tissue cell pool composition as well as to a decreased pancreatic secretion of the anabolic hormone insulin. These effects should be considered when interpreting results of experimental BMT in metabolic studies.


Subject(s)
Adipocytes/cytology , Bone Marrow Transplantation , Diet, High-Fat , Adipose Tissue, White/cytology , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Animals , C-Peptide/blood , Diet, Fat-Restricted , Eating , Energy Metabolism , Fatty Acids, Nonesterified/analysis , Feces/chemistry , Glucose Tolerance Test , Insulin/blood , Insulin/metabolism , Insulin Secretion , Macrophages/cytology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/prevention & control , Pancreas/metabolism , Pancreas/pathology , Triglycerides/analysis , Weight Gain
10.
Thromb Res ; 136(2): 474-80, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26153047

ABSTRACT

Activated coagulation factor XII (α-FXIIa) is able to bind to fibrin(ogen) and increases the density and stiffness of the fibrin clot. Conversely, proteins of the contact system and the fibrinolytic system show a high degree of homology and α-FXIIa can convert plasminogen into plasmin resulting in fibrin degradation. Therefore, we studied the contribution of α-FXIIa to overall clot stability and plasmin driven fibrinolysis in the absence and presence of tissue plasminogen activator (tPA). We observed that α-FXIIa directly converted plasminogen into plasmin and reduced clot lysis time at all tPA concentrations tested (15-1500 pM). Simultaneous assessment of plasmin generation (chromogenic substrate S-2251) and fibrin formation and degradation (absorbance at 405nm), showed an earlier onset of fibrinolysis and plasmin formation in the presence of α-FXIIa. Fibrinolysis of clots formed under flow conditions, revealed that incorporation of α-FXIIa accelerated clot breakdown (fluorescence release of labeled fibrin) by additional plasmin generation on top of formation by tPA. Scanning electron microscopy (SEM) revealed that the surface area pore size increased in the presence compared with the absence of α-FXIIa when fibrinolysis was initiated by the conversion of plasminogen with tPA during clot formation. α-FXIIa enhances fibrinolysis in the presence of plasminogen, irrespective of whether tPA was present during clot formation or was added afterwards to initiate fibrinolysis. We postulate that FXIIa first strengthens the clot structure during clot formation and thereafter contributes towards fibrinolysis.


Subject(s)
Blood Coagulation/physiology , Factor XIIa/metabolism , Fibrinolysin/metabolism , Fibrinolysis/physiology , Humans , Whole Blood Coagulation Time
SELECTION OF CITATIONS
SEARCH DETAIL
...