Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 419
Filter
1.
Front Immunol ; 14: 1174140, 2023.
Article in English | MEDLINE | ID: mdl-37638013

ABSTRACT

Introduction: The mechanism underlying radiation-induced gut microbiota dysbiosis is undefined. This study examined the effect of radiation on the intestinal Paneth cell α-defensin expression and its impact on microbiota composition and mucosal tissue injury and evaluated the radio-mitigative effect of human α-defensin 5 (HD5). Methods: Adult mice were subjected to total body irradiation, and Paneth cell α-defensin expression was evaluated by measuring α-defensin mRNA by RT-PCR and α-defensin peptide levels by mass spectrometry. Vascular-to-luminal flux of FITC-inulin was measured to evaluate intestinal mucosal permeability and endotoxemia by measuring plasma lipopolysaccharide. HD5 was administered in a liquid diet 24 hours before or after irradiation. Gut microbiota was analyzed by 16S rRNA sequencing. Intestinal epithelial junctions were analyzed by immunofluorescence confocal microscopy and mucosal inflammatory response by cytokine expression. Systemic inflammation was evaluated by measuring plasma cytokine levels. Results: Ionizing radiation reduced the Paneth cell α-defensin expression and depleted α-defensin peptides in the intestinal lumen. α-Defensin down-regulation was associated with the time-dependent alteration of gut microbiota composition, increased gut permeability, and endotoxemia. Administration of human α-defensin 5 (HD5) in the diet 24 hours before irradiation (prophylactic) significantly blocked radiation-induced gut microbiota dysbiosis, disruption of intestinal epithelial tight junction and adherens junction, mucosal barrier dysfunction, and mucosal inflammatory response. HD5, administered 24 hours after irradiation (treatment), reversed radiation-induced microbiota dysbiosis, tight junction and adherens junction disruption, and barrier dysfunction. Furthermore, HD5 treatment also prevents and reverses radiation-induced endotoxemia and systemic inflammation. Conclusion: These data demonstrate that radiation induces Paneth cell dysfunction in the intestine, and HD5 feeding prevents and mitigates radiation-induced intestinal mucosal injury, endotoxemia, and systemic inflammation.


Subject(s)
Endotoxemia , Radiation Injuries , alpha-Defensins , Humans , Adult , Animals , Mice , Paneth Cells , Dysbiosis , Endotoxemia/etiology , RNA, Ribosomal, 16S , Radiation Injuries/etiology , Cytokines , Inflammation
2.
Nutr Res ; 112: 20-29, 2023 04.
Article in English | MEDLINE | ID: mdl-36934524

ABSTRACT

Vitamin D insufficiency induces calcification disorder of bone or a decrease in bone mineral density, increasing the risk of fracture. Alkaline phosphatase (ALP) activity, a differentiation marker for intestinal epithelial cells, is regulated by vitamin D. It has also been suggested that ALP may prevent metabolic endotoxemia by dephosphorylating lipopolysaccharide. We hypothesized that vitamin D restriction and/or a high-fat diet influences ALP activity in each tissue and serum lipopolysaccharide concentrations and increases the risk of metabolic endotoxemia. Eleven-week-old female rats were divided into 4 groups: basic control diet (Cont.), basic control diet with vitamin D restriction (DR), high-fat diet (HF), and high-fat diet with vitamin D restriction (DRHF) groups. They were acclimated for 28 days. The results of 2-way analysis of variance showed that intestinal ALP activity, which may contribute to an improvement in phosphate/lipid metabolism and longevity, in the high-fat diet groups (HF and DRHF) was higher than in the low-fat diet groups (Cont. and DR). ALP activity in the vitamin D-restricted groups (DR and DRHF) was lower than in the vitamin D-sufficient groups (Cont. and HF). Furthermore, serum endotoxin concentrations were significantly higher in the high-fat diet groups (HF and DRHF) than in the low-fat diet groups (Cont. and DR). In the vitamin D-restricted groups (DR and DRHF), serum endotoxin concentrations were also significantly higher than in the vitamin D-sufficient groups (Cont. and HF). These results suggest that vitamin D restriction and/or a high-fat diet increases the risk of metabolic endotoxemia.


Subject(s)
Diet, High-Fat , Endotoxemia , Vitamin D , Animals , Female , Rats , Alkaline Phosphatase/metabolism , Diet, High-Fat/adverse effects , Endotoxemia/etiology , Endotoxins/blood , Lipopolysaccharides , Vitamins
3.
Int J Obes (Lond) ; 46(10): 1892-1900, 2022 10.
Article in English | MEDLINE | ID: mdl-35933445

ABSTRACT

AIMS/HYPOTHESIS: Translocation of bacterial debris from the gut causes metabolic endotoxemia (ME) that results in insulin resistance, and may be on the causal pathway to obesity-related type 2 diabetes. To guide interventions against ME we tested two hypothesised mechanisms for lipopolysaccharide (LPS) ingress: a leaky gut and chylomicron-associated transfer following a high-fat meal. METHODS: In lean women (n = 48; fat mass index (FMI) 9.6 kg/m2), women with obesity (n = 62; FMI 23.6 kg/m2) and women with obesity-diabetes (n = 38; FMI 24.9 kg/m2) we used the lactulose-mannitol dual-sugar permeability test (LM ratio) to assess gut integrity. Markers of ME (LPS, EndoCAb IgG and IgM, IL-6, CD14 and lipoprotein binding protein) were assessed at baseline, 2 h and 5 h after a standardised 49 g fat-containing mixed meal. mRNA expression of markers of inflammation, macrophage activation and lipid metabolism were measured in peri-umbilical adipose tissue (AT) biopsies. RESULTS: The LM ratio did not differ between groups. LPS levels were 57% higher in the obesity-diabetes group (P < 0.001), but, contrary to the chylomicron transfer hypothesis, levels significantly declined following the high-fat challenge. EndoCAb IgM was markedly lower in women with obesity and women with obesity-diabetes. mRNA levels of inflammatory markers in adipose tissue were consistent with the prior concept that fat soluble LPS in AT attracts and activates macrophages. CONCLUSIONS/INTERPRETATION: Raised levels of LPS and IL-6 in women with obesity-diabetes and evidence of macrophage activation in adipose tissue support the concept of metabolic endotoxemia-mediated inflammation, but we found no evidence for abnormal gut permeability or chylomicron-associated post-prandial translocation of LPS. Instead, the markedly lower EndoCAb IgM levels indicate a failure in sequestration and detoxification.


Subject(s)
Diabetes Mellitus, Type 2 , Endotoxemia , Chylomicrons , Diabetes Mellitus, Type 2/complications , Endotoxemia/etiology , Female , Gambia , Humans , Immunoglobulin G , Immunoglobulin M , Inflammation/metabolism , Interleukin-6 , Lactulose , Lipopolysaccharides/metabolism , Lipoproteins/metabolism , Mannitol , Obesity/metabolism , RNA, Messenger
4.
Tissue Eng Regen Med ; 19(2): 403-415, 2022 04.
Article in English | MEDLINE | ID: mdl-35122584

ABSTRACT

BACKGROUND: Endotoxemia is related to worse clinical outcomes in acute liver failure (ALF), but its management remains unsatisfactory. In this study, we aimed to assess whether the application of bone marrow mesenchymal stem cells (BMSCs) could eliminate endotoxemia and protect rats against ALF induced by thioacetamide (TAA). METHODS: BMSCs were isolated from rats and identified by the specific morphology, differentiation potential, and surface markers. The optimal dose of TAA for this study was explored and TAA-induced ALF rats were randomized to three groups: the normal control group (Saline), ALF group (TAA + Saline), and BMSCs-treated group (TAA + BMSCs). The intestinal migration and differentiation of BMSCs was tracked in vivo, and intestinal permeability, endotoxin and inflammatory cytokines, histology, and mortality were analyzed. Moreover, we added the inhibitor of the PI3K/AKT/mTOR signaling pathway into the co-culture system of BMSCs with enterocytes and then performed CK and Villin expression experiments to assess the role of PI3K/AKT/mTOR signal pathway in the intestinal differentiation of BMSCs. RESULTS: BMSCs migrated to the intestinal injury sites and differentiated into enterocytes, intestinal permeability was decreased compared with the ALF group. The higher expression of endotoxin and inflammatory cytokines were reversed after BMSCs transplantation in rats with ALF. Mortality and intestinal lesion were significantly decreased. Blocking the PI3K/AKT/mTOR signal pathway inhibited BMSCs' intestinal differentiation in vitro. CONCLUSION: BMSCs can eliminate endotoxemia and reduce mortality in rats with ALF, and the PI3K/AKT/mTOR signal pathway is involved in intestinal differentiation. BMSCs transplantation could be a potential candidate for the treatment of endotoxemia in ALF.


Subject(s)
Endotoxemia , Liver Failure, Acute , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Rats , Bone Marrow Cells , Endotoxemia/etiology , Endotoxemia/metabolism , Endotoxemia/therapy , Liver Failure, Acute/chemically induced , Liver Failure, Acute/metabolism , Liver Failure, Acute/therapy , Mesenchymal Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rats, Sprague-Dawley , Thioacetamide/metabolism , Thioacetamide/toxicity
5.
Int J Mol Sci ; 23(3)2022 Jan 19.
Article in English | MEDLINE | ID: mdl-35163007

ABSTRACT

Mechanical ventilation (MV) is essential for patients with sepsis-related respiratory failure but can cause ventilator-induced diaphragm dysfunction (VIDD), which involves diaphragmatic myofiber atrophy and contractile inactivity. Mitochondrial DNA, oxidative stress, mitochondrial dynamics, and biogenesis are associated with VIDD. Hypoxia-inducible factor 1α (HIF-1α) is crucial in the modulation of diaphragm immune responses. The mechanism through which HIF-1α and mitochondria affect sepsis-related diaphragm injury is unknown. We hypothesized that MV with or without endotoxin administration would aggravate diaphragmatic and mitochondrial injuries through HIF-1α. C57BL/6 mice, either wild-type or HIF-1α-deficient, were exposed to MV with or without endotoxemia for 8 h. MV with endotoxemia augmented VIDD and mitochondrial damage, which presented as increased oxidative loads, dynamin-related protein 1 level, mitochondrial DNA level, and the expressions of HIF-1α and light chain 3-II. Furthermore, disarrayed myofibrils; disorganized mitochondria; increased autophagosome numbers; and substantially decreased diaphragm contractility, electron transport chain activities, mitofusin 2, mitochondrial transcription factor A, peroxisome proliferator activated receptor-γ coactivator-1α, and prolyl hydroxylase domain 2 were observed (p < 0.05). Endotoxin-stimulated VIDD and mitochondrial injuries were alleviated in HIF-1α-deficient mice (p < 0.05). Our data revealed that endotoxin aggravated MV-induced diaphragmatic dysfunction and mitochondrial damages, partially through the HIF-1α signaling pathway.


Subject(s)
Diaphragm/injuries , Endotoxemia/therapy , Endotoxins/adverse effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mitochondria/metabolism , Respiration, Artificial/adverse effects , Animals , Diaphragm/metabolism , Diaphragm/physiopathology , Disease Models, Animal , Endotoxemia/etiology , Endotoxemia/metabolism , Gene Knockout Techniques , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Muscle Contraction , Oxidative Stress , Signal Transduction
6.
Int J Mol Sci ; 23(3)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35163089

ABSTRACT

Lipopolysaccharide (LPS)-induced endotoxemia induces an acute systemic inflammatory response that mimics some important features of sepsis, the disease with the highest mortality rate worldwide. In this work, we have analyzed a murine model of endotoxemia based on a single intraperitoneal injection of 5 mg/kg of LPS. We took advantage of galectin-3 (Gal3) knockout mice and found that the absence of Gal3 decreased the mortality rate oflethal endotoxemia in the first 80 h after the administration of LPS, along with a reduction in the tissular damage in several organs measured by electron microscopy. Using flow cytometry, we demonstrated that, in control conditions, peripheral immune cells, especially monocytes, exhibited high levels of Gal3, which were early depleted in response to LPS injection, thus suggesting Gal3 release under endotoxemia conditions. However, serum levels of Gal3 early decreased in response to LPS challenge (1 h), an indication that Gal3 may be extravasated to peripheral organs. Indeed, analysis of Gal3 in peripheral organs revealed a robust up-regulation of Gal3 36 h after LPS injection. Taken together, these results demonstrate the important role that Gal3 could play in the development of systemic inflammation, a well-established feature of sepsis, thus opening new and promising therapeutic options for these harmful conditions.


Subject(s)
Disease Models, Animal , Endotoxemia/pathology , Galectin 3/physiology , Inflammation/pathology , Lipopolysaccharides/toxicity , Macrophages, Peritoneal/immunology , Animals , Endotoxemia/etiology , Endotoxemia/metabolism , Inflammation/etiology , Inflammation/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
Am J Respir Cell Mol Biol ; 66(2): 183-195, 2022 02.
Article in English | MEDLINE | ID: mdl-34706199

ABSTRACT

TLR4 signaling via endotoxemia in macrophages promotes macrophage transition to the inflammatory phenotype through NLRP3 inflammasome activation. This transition event has the potential to trigger acute lung injury (ALI). However, relatively little is known about the regulation of NLRP3 and its role in the pathogenesis of ALI. Here we interrogated the signaling pathway activated by CD38, an ectoenzyme expressed in macrophages, in preventing ALI through suppressing NLRP3 activation. Wild-type and Cd38-knockout (Cd38-/-) mice were used to assess inflammatory lung injury, and isolated macrophages were used to delineate underlying TLR4 signaling pathway. We showed that CD38 suppressed TLR4 signaling in macrophages by inhibiting Bruton's tyrosine kinase (Btk) through the recruitment of Src homology-2 domain containing protein tyrosine phosphatase-2 (SHP2) and resulting in the dephosphorylation of activated Btk. Cd38-/- mice show enhanced lung polymorphonuclear leukocyte extravasation and severe lung injury. LPS- or polymicrobial sepsis-induced mortality in Cd38-/- mice were markedly augmented compared with wild types. CD38 in macrophages functioned by inhibiting Btk activation through activation of SHP2 and resulting dephosphorylation of Btk, and thereby preventing activation of downstream targets NF-κB and NLRP3. Cd38-/- macrophages displayed markedly increased activation of Btk, NF-κB, and NLRP3, whereas in vivo administration of the Btk inhibitor ibrutinib (a Food and Drug Administration-approved drug) prevented augmented TLR4-induced inflammatory lung injury seen in Cd38-/- mice. Our findings together show upregulation of CD38 activity and inhibition of Btk activation downstream of TLR4 activation as potential strategies to prevent endotoxemic ALI.


Subject(s)
ADP-ribosyl Cyclase 1/physiology , Acute Lung Injury/prevention & control , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Endotoxemia/prevention & control , Inflammasomes/drug effects , Macrophages/drug effects , Membrane Glycoproteins/physiology , Piperidines/pharmacology , Acute Lung Injury/etiology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Adenine/pharmacology , Agammaglobulinaemia Tyrosine Kinase/genetics , Agammaglobulinaemia Tyrosine Kinase/metabolism , Animals , Endotoxemia/etiology , Endotoxemia/metabolism , Endotoxemia/pathology , Female , Inflammasomes/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction
8.
Blood Purif ; 51(1): 47-54, 2022.
Article in English | MEDLINE | ID: mdl-33857940

ABSTRACT

INTRODUCTION: Uncontrolled systemic inflammation may occur in severe coronavirus disease 19 (COVID-19). We have previously shown that endotoxemia, presumably from the gut, may complicate COVID-19. However, the role of endotoxin adsorbent (EA) therapy to mitigate organ dysfunction in COVID-19 has not been explored. METHODS: We conducted a retrospective observational study in COVID-19 patients who received EA therapy at the King Chulalongkorn Memorial Hospital, Bangkok, Thailand, between March 13 and April 17, 2020. Relevant clinical and laboratory data were collected by inpatient chart review. RESULTS: Among 147 hospitalized COVID-19 patients, 6 patients received EA therapy. All of the 6 patients had severe COVID-19 infection with acute respiratory distress syndrome (ARDS). Among these, 5 of them were mechanically ventilated and 4 had complications of secondary bacterial infection. The endotoxin activity assay (EAA) results of pre-EA therapy ranged from 0.47 to 2.79. The choices of EA therapy were at the discretion of attending physicians. One patient was treated with oXiris® along with continuous renal replacement therapy, and the others received polymyxin B hemoperfusion sessions. All patients have survived and were finally free from the mechanical ventilation as well as had improvement in PaO2/FiO2 ratio and decreased EAA level after EA therapy. CONCLUSIONS: We demonstrated the clinical improvement of severe COVID-19 patients with elevated EAA level upon receiving EA therapy. However, the benefit of EA therapy in COVID-19 ARDS is still unclear and needs to be elucidated with randomized controlled study.


Subject(s)
COVID-19/therapy , Endotoxemia/therapy , Hemoperfusion/methods , SARS-CoV-2 , Acute Kidney Injury/etiology , Acute Kidney Injury/therapy , Adsorption , COVID-19/complications , Critical Care/methods , Endotoxemia/etiology , Female , Heparin/administration & dosage , Humans , Male , Membranes, Artificial , Middle Aged , Polymyxin B/administration & dosage , Renal Replacement Therapy , Respiratory Distress Syndrome/etiology , Retrospective Studies , Treatment Outcome
9.
Life Sci ; 288: 120153, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34801513

ABSTRACT

AIMS: To characterize neuroinflammatory and gut dysbiosis signatures that accompany exaggerated exercise fatigue and cognitive/mood deficits in a mouse model of Gulf War Illness (GWI). METHODS: Adult male C57Bl/6N mice were exposed for 28 d (5 d/wk) to pyridostigmine bromide (P.O.) at 6.5 mg/kg/d, b.i.d. (GW1) or 8.7 mg/kg/d, q.d. (GW2); topical permethrin (1.3 mg/kg), topical N,N-diethyl-meta-toluamide (33%) and restraint stress (5 min). Animals were phenotypically evaluated as described in an accompanying article [124] and sacrificed at 6.6 months post-treatment (PT) to allow measurement of brain neuroinflammation/neuropathic pain gene expression, hippocampal glial fibrillary acidic protein, brain Interleukin-6, gut dysbiosis and serum endotoxin. KEY FINDINGS: Compared to GW1, GW2 showed a more intense neuroinflammatory transcriptional signature relative to sham stress controls. Interleukin-6 was elevated in GW2 and astrogliosis in hippocampal CA1 was seen in both GW groups. Beta-diversity PCoA using weighted Unifrac revealed that gut microbial communities changed after exposure to GW2 at PT188. Both GW1 and GW2 displayed systemic endotoxemia, suggesting a gut-brain mechanism underlies the neuropathological signatures. Using germ-free mice, probiotic supplementation with Lactobacillus reuteri produced less gut permeability than microbiota transplantation using GW2 feces. SIGNIFICANCE: Our findings demonstrate that GW agents dose-dependently induce differential neuropathology and gut dysbiosis associated with cognitive, exercise fatigue and mood GWI phenotypes. Establishment of a comprehensive animal model that recapitulates multiple GWI symptom domains and neuroinflammation has significant implications for uncovering pathophysiology, improving diagnosis and treatment for GWI.


Subject(s)
Cognitive Dysfunction/pathology , Dysbiosis/pathology , Fatigue/pathology , Gastrointestinal Microbiome , Neuroinflammatory Diseases/pathology , Persian Gulf Syndrome/drug therapy , Physical Conditioning, Animal , Pyridostigmine Bromide/toxicity , Animals , Biomarkers/analysis , Cholinesterase Inhibitors/administration & dosage , Cholinesterase Inhibitors/toxicity , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Dysbiosis/etiology , Dysbiosis/metabolism , Endotoxemia/etiology , Endotoxemia/metabolism , Endotoxemia/pathology , Fatigue/etiology , Fatigue/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gliosis/etiology , Gliosis/metabolism , Gliosis/pathology , Male , Mice , Mice, Inbred C57BL , Neuralgia/etiology , Neuralgia/metabolism , Neuralgia/pathology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Pyridostigmine Bromide/administration & dosage
10.
Int J Mol Sci ; 22(21)2021 Nov 03.
Article in English | MEDLINE | ID: mdl-34769369

ABSTRACT

Competition for the amino acid arginine by endothelial nitric-oxide synthase (NOS3) and (pro-)inflammatory NO-synthase (NOS2) during endotoxemia appears essential in the derangement of the microcirculatory flow. This study investigated the role of NOS2 and NOS3 combined with/without citrulline supplementation on the NO-production and microcirculation during endotoxemia. Wildtype (C57BL6/N background; control; n = 36), Nos2-deficient, (n = 40), Nos3-deficient (n = 39) and Nos2/Nos3-deficient mice (n = 42) received a continuous intravenous LPS infusion alone (200 µg total, 18 h) or combined with L-citrulline (37.5 mg, last 6 h). The intestinal microcirculatory flow was measured by side-stream dark field (SDF)-imaging. The jejunal intracellular NO production was quantified by in vivo NO-spin trapping combined with electron spin-resonance (ESR) spectrometry. Amino-acid concentrations were measured by high-performance liquid chromatography (HPLC). LPS infusion decreased plasma arginine concentration in control and Nos3-/- compared to Nos2-/- mice. Jejunal NO production and the microcirculation were significantly decreased in control and Nos2-/- mice after LPS infusion. No beneficial effects of L-citrulline supplementation on microcirculatory flow were found in Nos3-/- or Nos2-/-/Nos3-/- mice. This study confirms that L-citrulline supplementation enhances de novo arginine synthesis and NO production in mice during endotoxemia with a functional NOS3-enzyme (control and Nos2-/- mice), as this beneficial effect was absent in Nos3-/- or Nos2-/-/Nos3-/- mice.


Subject(s)
Arginine/metabolism , Citrulline/administration & dosage , Endotoxemia/pathology , Microcirculation , NADPH Oxidase 2/physiology , NADPH Oxidases/physiology , Nitric Oxide/metabolism , Animals , Endotoxemia/drug therapy , Endotoxemia/etiology , Intestines/drug effects , Intestines/metabolism , Intestines/pathology , Jejunum/drug effects , Jejunum/metabolism , Jejunum/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Int J Mol Sci ; 22(17)2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34502470

ABSTRACT

Bacterial endotoxin is a potent inflammatory antigen abundant in the human intestine. Endotoxins circulate in the blood at low concentrations in all healthy individuals. Elevated levels of circulatory endotoxins may cause inflammation with the development of chronic disease, either affecting metabolism, neurological disease, or resistance to viral and bacterial infections. The most important endotoxin is LPS, being a superantigen. In this narrative review, the effect of various food components to postprandially elevate circulating LPS and inflammatory markers is described. There is evidence that the intake of food enriched in fat, in particular saturated fat, may elevate LPS and pro-inflammatory markers. This occurs in both normal-weight and obese subjects. In obese subjects, inflammatory markers are already elevated before meal consumption. The importance of food choice for endotoxemia and inflammatory response is discussed.


Subject(s)
Dietary Fats , Endotoxemia/blood , Food Contamination , Lipopolysaccharides/toxicity , Obesity/blood , Biomarkers/blood , Endotoxemia/etiology , Humans , Inflammation/blood , Inflammation/etiology
12.
Cell Rep ; 36(11): 109691, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525353

ABSTRACT

Lipopolysaccharides (LPSs) can promote metabolic endotoxemia, which is considered inflammatory and metabolically detrimental based on Toll-like receptor (TLR)4 agonists, such as Escherichia coli-derived LPS. LPSs from certain bacteria antagonize TLR4 yet contribute to endotoxemia measured by endotoxin units (EUs). We found that E. coli LPS impairs gut barrier function and worsens glycemic control in mice, but equal doses of LPSs from other bacteria do not. Matching the LPS dose from R. sphaeroides and E. coli by EUs reveals that only E. coli LPS promotes dysglycemia and adipose inflammation, delays intestinal glucose absorption, and augments insulin and glucagon-like peptide (GLP)-1 secretion. Metabolically beneficial endotoxemia promoted by R. sphaeroides LPS counteracts dysglycemia caused by an equal dose of E. coli LPS and improves glucose control in obese mice. The concept of metabolic endotoxemia should be expanded beyond LPS load to include LPS characteristics, such as lipid A acylation, which dictates the effect of metabolic endotoxemia.


Subject(s)
Endotoxemia/etiology , Intestines/drug effects , Lipopolysaccharides/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Blood Glucose/analysis , Body Weight/drug effects , Endotoxemia/metabolism , Escherichia coli/metabolism , Glucagon-Like Peptide 1/blood , Glucose/metabolism , Insulin/blood , Intestines/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Obesity/pathology , Peptidoglycan/pharmacology , Rhodobacter sphaeroides/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/metabolism
13.
Nutrients ; 13(8)2021 Aug 15.
Article in English | MEDLINE | ID: mdl-34444955

ABSTRACT

Diet and dietary components have profound effects on the composition of the gut microbiota and are among the most important contributors to the alteration in bacterial flora. This review examines the effects the "Western", "plant-based", "high-fat", "medical ketogenic", and "Mediterranean" diets have on the composition of the gut microbiota in both mice and human subjects. We show that specific dietary components that are commonly found in the "plant-based" and "Mediterranean" diet play a role in shifting the microbial composition. This review further evaluates the bacterial metabolites that are associated with diet, and their role in systemic inflammation and metabolic endotoxemia. Furthermore, the associations between diet/dietary components and altering bacterial composition, may lead to potential therapeutic targets for type II diabetes, obesity, and inflammatory diseases.


Subject(s)
Diet/adverse effects , Gastrointestinal Microbiome/physiology , Nutritional Physiological Phenomena , Animals , Diet/methods , Diet, High-Fat/adverse effects , Diet, Ketogenic/adverse effects , Diet, Mediterranean/adverse effects , Diet, Vegetarian/adverse effects , Diet, Western/adverse effects , Endotoxemia/etiology , Endotoxemia/microbiology , Humans , Inflammation/etiology , Inflammation/microbiology , Mice
14.
Front Immunol ; 12: 701275, 2021.
Article in English | MEDLINE | ID: mdl-34349763

ABSTRACT

Metabolic endotoxemia has been suggested to play a role in the pathophysiology of metaflammation, insulin-resistance and ultimately type-2 diabetes mellitus (T2DM). The role of endogenous antimicrobial peptides (AMPs), such as the cathelicidin LL-37, in T2DM is unknown. We report here for the first time that patients with T2DM compared to healthy volunteers have elevated plasma levels of LL-37. In a reverse-translational approach, we have investigated the effects of the AMP, peptide 19-2.5, in a murine model of high-fat diet (HFD)-induced insulin-resistance, steatohepatitis and T2DM. HFD-fed mice for 12 weeks caused obesity, an impairment in glycemic regulations, hypercholesterolemia, microalbuminuria and steatohepatitis, all of which were attenuated by Peptide 19-2.5. The liver steatosis caused by feeding mice a HFD resulted in the activation of nuclear factor kappa light chain enhancer of activated B cells (NF-ĸB) (phosphorylation of inhibitor of kappa beta kinase (IKK)α/ß, IκBα, translocation of p65 to the nucleus), expression of NF-ĸB-dependent protein inducible nitric oxide synthase (iNOS) and activation of the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome, all of which were reduced by Peptide 19-2.5. Feeding mice, a HFD also resulted in an enhanced expression of the lipid scavenger receptor cluster of differentiation 36 (CD36) secondary to activation of extracellular signal-regulated kinases (ERK)1/2, both of which were abolished by Peptide 19-2.5. Taken together, these results demonstrate that the AMP, Peptide 19-2.5 reduces insulin-resistance, steatohepatitis and proteinuria. These effects are, at least in part, due to prevention of the expression of CD36 and may provide further evidence for a role of metabolic endotoxemia in the pathogenesis of metaflammation and ultimately T2DM. The observed increase in the levels of the endogenous AMP LL-37 in patients with T2DM may serve to limit the severity of the disease.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Antimicrobial Peptides/pharmacology , Diabetes Mellitus, Type 2/metabolism , Inflammation , Lipopolysaccharides/antagonists & inhibitors , Animals , Diet, High-Fat/adverse effects , Endotoxemia/etiology , Endotoxemia/metabolism , Humans , Inflammation/etiology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Cathelicidins
15.
Shock ; 56(2): 268-277, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34276040

ABSTRACT

ABSTRACT: Leukocyte Nox2 is recognized to have a fundamental microbicidal function in sepsis but the specific role of Nox2 in endothelial cells (EC) remains poorly elucidated. Here, we tested the hypothesis that endothelial Nox2 participates in the pathogenesis of systemic inflammation and hypotension induced by LPS. LPS was injected intravenously in mice with Tie2-targeted deficiency or transgenic overexpression of Nox2. Mice with Tie2-targeted Nox2 deficiency had increased circulating levels of TNF-α, enhanced numbers of neutrophils trapped in lungs, and aggravated hypotension after LPS injection, as compared to control LPS-injected animals. In contrast, Tie2-driven Nox2 overexpression attenuated inflammation and prevented the hypotension induced by LPS. Because Tie2-Cre targets both EC and myeloid cells we generated bone marrow chimeric mice with Nox2 deletion restricted to leukocytes or ECs. Mice deficient in Nox2 either in leukocytes or ECs had reduced LPS-induced neutrophil trapping in the lungs and lower plasma TNF-α levels as compared to control LPS-injected mice. However, the pronounced hypotensive response to LPS was present only in mice with EC-specific Nox2 deletion. Experiments in vitro with human vein or aortic endothelial cells (HUVEC and HAEC, respectively) treated with LPS revealed that EC Nox2 controls NF-κB activation and the transcription of toll-like receptor 4 (TLR4), which is the recognition receptor for LPS. In conclusion, these results suggest that endothelial Nox2 limits NF-κB activation and TLR4 expression, which in turn attenuates the severity of hypotension and systemic inflammation induced by LPS.


Subject(s)
Endothelial Cells/physiology , Endotoxemia/etiology , Hypotension/etiology , Inflammation/etiology , NADPH Oxidase 2/physiology , Toll-Like Receptor 4/physiology , Animals , Male , Mice , Mice, Inbred C57BL
16.
Front Immunol ; 12: 658404, 2021.
Article in English | MEDLINE | ID: mdl-34163471

ABSTRACT

Chronic kidney disease induces disruption of the intestinal epithelial barrier, leading to gut bacterial translocation. Here, we appreciated bacterial translocation by analyzing circulating lipopolysaccharides (LPS) using two methods, one measuring only active free LPS, and the other quantifying total LPS as well as LPS lipid A carbon chain length. This was done in end-stage renal disease (ESRD) patients and healthy volunteers (HV). We observed both higher LPS concentration in healthy volunteers and significant differences in composition of translocated LPS based on lipid A carbon chain length. Lower LPS activity to mass ratio and higher concentration of high-density lipoproteins were found in HV, suggesting a better plasma capacity to neutralize LPS activity. Higher serum concentrations of soluble CD14 and pro-inflammatory cytokines in ESRD patients confirmed this hypothesis. To further explore whether chronic inflammation in ESRD patients could be more related to LPS composition rather than its quantity, we tested the effect of HV and patient sera on cytokine secretion in monocyte cultures. Sera with predominance of 14-carbon chain lipid A-LPS induced higher secretion of pro-inflammatory cytokines than those with predominance of 18-carbon chain lipid A-LPS. TLR4 or LPS antagonists decreased LPS-induced cytokine production by monocytes, demonstrating an LPS-specific effect. Thereby, septic inflammation observed in ESRD patients may be not related to higher bacterial translocation, but to reduced LPS neutralization capacity and differences in translocated LPS subtypes.


Subject(s)
Bacterial Translocation , Disease Susceptibility , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/therapy , Adult , Aged , Biomarkers , Case-Control Studies , Comorbidity , Cytokines/blood , Disease Management , Disease Susceptibility/immunology , Endotoxemia/diagnosis , Endotoxemia/etiology , Female , Humans , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/diagnosis , Kidney Transplantation , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Male , Middle Aged , Monocytes/immunology , Monocytes/metabolism
17.
Nutr Res ; 91: 26-35, 2021 07.
Article in English | MEDLINE | ID: mdl-34130208

ABSTRACT

Current evidence suggests that high fructose intake results in gut dysbiosis, leading to endotoxemia and NAFLD onset. Thus, the hypothesis of the study was that an enhanced Proteobacteria proportion in the cecal microbiota could be the most prominent trigger of NAFLD through enhanced endotoxin (LPS) in adult high-fructose-fed C57BL/6 mice. Male C57BL/6 mice received a control diet (n = 10, C: 76% of energy as carbohydrates, 0% as fructose) or high-fructose diet (n = 10, HFRU: 76% of energy as carbohydrate, 50% as fructose) for 12 weeks. Outcomes included biochemical analyses, 16S rDNA PCR amplification, hepatic stereology, and RT-qPCR. The groups showed similar body masses during the whole experiment. However, the HFRU group showed greater water intake and blood pressure than the C group. The HFRU group showed a significantly lower amount of Bacteroidetes and a predominant rise in Proteobacteria, implying increased LPS. The HFRU group also showed enhanced de novo lipogenesis (Chrebp expression), while beta-oxidation was decreased (Ppar-alpha expression). These results agree with the deposition of fat droplets within hepatocytes and the enhanced hepatic triacylglycerol concentrations, as observed in the photomicrographs, where the HFRU group had a higher volume density of steatosis than the C group. Thus, we confirmed that a rise in the Proteobacteria phylum proportion was the most prominent alteration in gut-liver axis-induced hepatic steatosis in HFRU-fed C57BL/6 mice. Gut dysbiosis and fatty liver were observed even in the absence of overweight in this dietary adult mouse model.


Subject(s)
Diet/adverse effects , Dysbiosis/microbiology , Fructose/adverse effects , Gastrointestinal Microbiome , Liver , Non-alcoholic Fatty Liver Disease/microbiology , Proteobacteria/growth & development , Animals , Body Weight , Cecum/microbiology , Dietary Sugars/adverse effects , Disease Models, Animal , Dysbiosis/etiology , Endotoxemia/etiology , Endotoxemia/microbiology , Feeding Behavior , Lipid Metabolism , Lipopolysaccharides , Liver/metabolism , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Triglycerides/metabolism
18.
Shock ; 56(4): 537-543, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34172613

ABSTRACT

INTRODUCTION: Early diagnosis and treatment can reduce the risk of organ failure and mortality in systemic inflammatory conditions. Heart rate variability (HRV) has potential for early identification of the onset of systemic inflammation, as it may detect changes in sympathetic nervous system activity resulting from the developing inflammatory response before clinical signs appear. With the use of new methodologies, we investigated the onset and kinetics of HRV changes as well as several inflammatory parameters and symptoms during experimental human endotoxemia, a model of systemic inflammation in humans in vivo. PATIENTS AND METHODS: Healthy volunteers were intravenously administered LPS (n = 15) or placebo (n = 15). HRV was determined using a wireless wearable device, and parameters low to high frequency (LF:HF) ratio, root mean square of the successive differences (RMSSD), and standard deviation of normal-to-normal R-R intervals (SDNN)were calculated through 1-min-rolling 6-min windows. Plasma cytokine levels and flu-like symptoms and vital signs were serially assessed. RESULTS: The increase in LF:HF ratio, reflecting sympathetic predominance, was more pronounced in the LPS group compared to the placebo group, with the difference becoming statistically significant 65 min following LPS administration (1.63 [1.42-1.83] vs. 1.28 [1.11-1.44], P = 0.005). Significant between-group differences in RMSSD and SDNN were observed from 127 to 140 min post-LPS administration onwards, respectively. Plasma cytokine levels showed significant between-group differences staring 60 min post-LPS. For symptom score, heart rate, temperature, and diastolic blood pressure, significant differences compared with the placebo group were observed at 90, 118, 120, and 124 min post-LPS, respectively. CONCLUSION: In a controlled human model of systemic inflammation, elevations in the LF:HF ratio followed very shortly after elevations in plasma cytokine levels and preceded onset of flu-like symptoms and alterations in vital signs. HRV may represent a promising non-invasive tool for early detection of a developing systemic inflammatory response.


Subject(s)
Electrocardiography, Ambulatory/instrumentation , Endotoxemia/diagnosis , Endotoxemia/physiopathology , Heart Rate/physiology , Sympathetic Nervous System/physiopathology , Wearable Electronic Devices , Cytokines/blood , Endotoxemia/etiology , Humans , Lipopolysaccharides , Male , Young Adult
19.
Epilepsia ; 62(6): 1472-1481, 2021 06.
Article in English | MEDLINE | ID: mdl-33893636

ABSTRACT

OBJECTIVE: Traumatic brain injury (TBI) may lead to the disruption of the intestinal barrier (IB), and to the escape of products of commensal gut bacteria, including lipopolysaccharide (LPS), into the bloodstream. We examined whether lateral fluid percussion injury (LFPI) and post-traumatic epilepsy (PTE) are associated with the increased intestinal permeability and endotoxemia, and whether these events in turn are associated with PTE. METHODS: LFPI was delivered to adult male Sprague-Dawley rats. Before, 1 week, and 7 months after LFPI, the IB permeability was examined by measuring plasma concentration of fluorescein isothiocyanate-labeled dextran (FD4) upon its enteral administration. Plasma LPS concentration was measured in the same animals, using enzyme-linked immunosorbent assay. PTE was examined 7 months after LFPI, with use of video-EEG (electroencephalography) monitoring. RESULTS: One week after LFPI, the IB disruption was detected in 14 of 17 and endotoxemia - in 10 of 17 rats, with a strong positive correlation between FD4 and LPS levels, and between plasma levels of each of the analytes and the severity of neuromotor deficit. Seven months after LFPI, IB disruption was detected in 13 of 15 and endotoxemia in 8 of 15 rats, with a strong positive correlation between plasma levels of the two analytes. Five of 15 LFPI rats developed PTE. Plasma levels of both FD4 and LPS were significantly higher in animals with PTE than among the animals without PTE. The analysis of seven rats, which were examined repeatedly at 1 week and at 7 months, confirmed that late IB disruption and endotoxemia were not due to lingering of impairments occurring shortly after LFPI. SIGNIFICANCE: LFPI leads to early and remote disruption of IB and a secondary endotoxemia. Early and late perturbations may occur in different subjects. Early changes reflect the severity of acute post-traumatic motor dysfunction, whereas late changes are associated with PTE.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Endotoxemia/physiopathology , Epilepsy, Post-Traumatic/physiopathology , Intestines/physiopathology , Animals , Brain Injuries, Traumatic/complications , Dextrans , Electroencephalography , Endotoxemia/etiology , Epilepsy, Post-Traumatic/complications , Fluorescein-5-isothiocyanate/analogs & derivatives , Lipopolysaccharides/blood , Male , Permeability , Rats , Rats, Sprague-Dawley
20.
Front Immunol ; 12: 609319, 2021.
Article in English | MEDLINE | ID: mdl-33679744

ABSTRACT

Endotoxemia is a severe inflammation response induced by infection especially bacterial endotoxin translocation, which severely increases mortality in combination with acute colon injury. Bromodomain-containing protein 4 (BRD4) is an important Bromo and Extra-Terminal (BET) protein to participate in inflammatory responses. However, it is still unknown about the specific connection between BRD4 and inflammation-related pyroptosis in endotoxemia colon. Here, through evaluating the mucous morphology and the expression of tight junction proteins such as occludin and ZO1, we found the upregulation of BRD4 in damaged colon with poor tight junction in an endotoxemia mouse model induced by lipopolysaccharides (LPS). Firstly, the BRD4 inhibitor JQ1 was used to effectively protect colon tight junction in endotoxemia. As detected, high levels of pro-inflammation cytokines IL6, IL1ß and IL18 in endotoxemia colon were reversed by JQ1 pretreatment. In addition, JQ1 injection reduced endotoxemia-induced elevation of the phosphorylated NF κB and NLRP3/ASC/caspase 1 inflammasome complex in colon injury. Furthermore, activated pyroptosis markers gasdermins in endotoxemia colon were also blocked by JQ1 pretreatment. Together, our data indicate that BRD4 plays a critical role in regulating pyroptosis-related colon injury induced by LPS, and JQ1 as a BRD4 inhibitors can effectively protect colon from endotoxemia-induced inflammation injury.


Subject(s)
Azepines/pharmacology , Colitis/metabolism , Nuclear Proteins/antagonists & inhibitors , Pyroptosis/drug effects , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Animals , Biomarkers , Colitis/drug therapy , Colitis/etiology , Colitis/pathology , Disease Models, Animal , Endotoxemia/complications , Endotoxemia/etiology , Immunohistochemistry , Inflammasomes/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lipopolysaccharides/adverse effects , Male , Mice , Phosphorylation , Tight Junctions/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...