Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Gut Microbes ; 16(1): 2370616, 2024.
Article in English | MEDLINE | ID: mdl-38961712

ABSTRACT

Amino acids, metabolized by host cells as well as commensal gut bacteria, have signaling effects on host metabolism. Oral supplementation of the essential amino acid histidine has been shown to exert metabolic benefits. To investigate whether dietary histidine aids glycemic control, we performed a case-controlled parallel clinical intervention study in participants with type 2 diabetes (T2D) and healthy controls. Participants received oral histidine for seven weeks. After 2 weeks of histidine supplementation, the microbiome was depleted by antibiotics to determine the microbial contribution to histidine metabolism. We assessed glycemic control, immunophenotyping of peripheral blood mononucelar cells (PBMC), DNA methylation of PBMCs and fecal gut microbiota composition. Histidine improves several markers of glycemic control, including postprandial glucose levels with a concordant increase in the proportion of MAIT cells after two weeks of histidine supplementation. The increase in MAIT cells was associated with changes in gut microbial pathways such as riboflavin biosynthesis and epigenetic changes in the amino acid transporter SLC7A5. Associations between the microbiome and MAIT cells were replicated in the MetaCardis cohort. We propose a conceptual framework for how oral histidine may affect MAIT cells via altered gut microbiota composition and SLC7A5 expression in MAIT cells directly and thereby influencing glycemic control. Future studies should focus on the role of flavin biosynthesis intermediates and SLC7A5 modulation in MAIT cells to modulate glycemic control.


Subject(s)
Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Histidine , Mucosal-Associated Invariant T Cells , Humans , Histidine/metabolism , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/immunology , Gastrointestinal Microbiome/drug effects , Middle Aged , Male , Female , Mucosal-Associated Invariant T Cells/immunology , Mucosal-Associated Invariant T Cells/metabolism , Glycemic Control , Dietary Supplements , Case-Control Studies , Feces/microbiology , Blood Glucose/metabolism , Aged , Adult , Bacteria/classification , Bacteria/metabolism , Bacteria/genetics , Administration, Oral , DNA Methylation
2.
Antioxidants (Basel) ; 12(10)2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37891903

ABSTRACT

The leading cause of mortality from SARS-CoV-2 is an exaggerated host immune response, triggering cytokine storms, multiple organ failure and death. Current drug- and vaccine-based therapies are of limited efficacy against novel viral variants. Infrared therapy is a non-invasive and safe method that has proven effective against inflammatory conditions for over 100 years. However, its mechanism of action is poorly understood and has not received widespread acceptance. We herein investigate whether near-infrared (NIR) light exposure in human primary alveolar and macrophage cells could downregulate inflammatory cytokines triggered by the SARS-CoV-2 spike (S) protein or lipopolysaccharide (LPS), and via what underlying mechanism. Our results showed a dramatic reduction in pro-inflammatory cytokines within days of NIR light treatment, while anti-inflammatory cytokines were upregulated. Mechanistically, NIR light stimulated mitochondrial metabolism, induced transient bursts in reactive oxygen species (ROS) and activated antioxidant gene transcription. These, in turn, downregulated ROS and inflammatory cytokines. A causal relationship was shown between the induction of cellular ROS by NIR light exposure and the downregulation of inflammatory cytokines triggered by SARS-CoV-2 S. If confirmed by clinical trials, this method would provide an immediate defense against novel SARS-CoV-2 variants and other inflammatory infectious diseases.

3.
Gut ; 71(12): 2463-2480, 2022 12.
Article in English | MEDLINE | ID: mdl-35017197

ABSTRACT

OBJECTIVES: Gut microbiota is a key component in obesity and type 2 diabetes, yet mechanisms and metabolites central to this interaction remain unclear. We examined the human gut microbiome's functional composition in healthy metabolic state and the most severe states of obesity and type 2 diabetes within the MetaCardis cohort. We focused on the role of B vitamins and B7/B8 biotin for regulation of host metabolic state, as these vitamins influence both microbial function and host metabolism and inflammation. DESIGN: We performed metagenomic analyses in 1545 subjects from the MetaCardis cohorts and different murine experiments, including germ-free and antibiotic treated animals, faecal microbiota transfer, bariatric surgery and supplementation with biotin and prebiotics in mice. RESULTS: Severe obesity is associated with an absolute deficiency in bacterial biotin producers and transporters, whose abundances correlate with host metabolic and inflammatory phenotypes. We found suboptimal circulating biotin levels in severe obesity and altered expression of biotin-associated genes in human adipose tissue. In mice, the absence or depletion of gut microbiota by antibiotics confirmed the microbial contribution to host biotin levels. Bariatric surgery, which improves metabolism and inflammation, associates with increased bacterial biotin producers and improved host systemic biotin in humans and mice. Finally, supplementing high-fat diet-fed mice with fructo-oligosaccharides and biotin improves not only the microbiome diversity, but also the potential of bacterial production of biotin and B vitamins, while limiting weight gain and glycaemic deterioration. CONCLUSION: Strategies combining biotin and prebiotic supplementation could help prevent the deterioration of metabolic states in severe obesity. TRIAL REGISTRATION NUMBER: NCT02059538.


Subject(s)
Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Obesity, Morbid , Vitamin B Complex , Humans , Mice , Animals , Prebiotics , Obesity, Morbid/surgery , Biotin/pharmacology , Vitamin B Complex/pharmacology , Mice, Inbred C57BL , Obesity/metabolism , Inflammation
4.
Stud Health Technol Inform ; 281: 53-57, 2021 May 27.
Article in English | MEDLINE | ID: mdl-34042704

ABSTRACT

Access to hospitals has been dramatically restricted during the COVID 19 pandemic. Indeed, due to the high risk of contamination by patients and by visitors, only essential visits and medical appointments have been authorized. Restricting hospital access to authorized visitors was an important logistic challenge. To deal with this challenge, our institution developed the ExpectingU app to facilitate patient authorization for medical appointments and for visitors to enter the hospital. This article analyzes different trends regarding medical appointments, visitors' invitations, support staff hired and COVID hospitalizations to demonstrate how the ExpectingU system has helped the hospital to maintain accessibility to the hospital. Results shows that our system has allowed us to maintain the hospital open for medical appointments and visits without creating bottlenecks.


Subject(s)
COVID-19 , Mobile Applications , Humans , Pandemics , SARS-CoV-2 , Visitors to Patients
5.
Diabetologia ; 64(1): 240-254, 2021 01.
Article in English | MEDLINE | ID: mdl-33125520

ABSTRACT

AIM/HYPOTHESIS: Altered adipose tissue secretory profile contributes to insulin resistance and type 2 diabetes in obesity. Preclinical studies have identified senescent cells as a cellular source of proinflammatory factors in adipose tissue of obese mice. In humans, potential links with obesity comorbidities are poorly defined. Here, we investigated adipose tissue senescent status and relationships with metabolic complications in human obesity. METHODS: The study includes a prospective cohort of 227 individuals with severe obesity. A photometric method was used to quantify senescence-associated ß-galactosidase (SA-ß-gal) activity in paired subcutaneous and omental adipose tissue biopsies obtained during gastric surgery. Gene and secretory profiling was performed in adipose tissue biopsies and in human primary pre-adipocytes in the presence or absence of senolytic drugs targeting senescent cells. Participants were phenotyped for anthropometric and bioclinical variables, metabolic complications and gastric surgery-induced improvement to address relationships with adipose tissue SA-ß-gal. RESULTS: SA-ß-gal activity was sevenfold higher in subcutaneous than in omental adipose tissue and not associated with BMI or chronological age. Several factors, including insulin-like growth factor binding protein 3 (IGFBP3), plasminogen activator inhibitor 1 (PAI1), C-C motif chemokine ligand 2 (CCL2) and IL-6, were upregulated in subcutaneous adipose tissue in relation with SA-ß-gal (p for linear trend across tertiles <0.05) and in pre-adipocytes cultured with inflammatory macrophage conditioned media. Senolytic treatment reduced SA-ß-gal staining and normalised these alterations. In the whole population, subcutaneous adipose tissue SA-ß-gal activity was positively associated with serum leptin, markers of insulin resistance and increased trunk fat mass. Metabolic complications, including type 2 diabetes and dyslipidaemia, were more prevalent in patients with high levels of SA-ß-gal, but improved with bariatric surgery whatever the initial adipose tissue senescent status. CONCLUSIONS/INTERPRETATION: This study highlights a phenotype of senescence in adipose tissue of severely obese individuals, which characterises prominently subcutaneous fat depots. Subcutaneous adipose tissue senescence is significantly linked to altered glucose metabolism and body fat distribution. Elimination of senescent cells through senolytic treatment could alleviate metabolic complications in severely obese people. Graphical abstract.


Subject(s)
Blood Glucose/analysis , Body Composition/physiology , Cellular Senescence/physiology , Obesity, Morbid/physiopathology , Subcutaneous Fat/enzymology , beta-Galactosidase/metabolism , Adipocytes/physiology , Bariatric Surgery , Biopsy , Cohort Studies , Female , Humans , Insulin Resistance , Male , Obesity, Morbid/metabolism , Obesity, Morbid/surgery , Prospective Studies , Subcutaneous Fat/pathology , Treatment Outcome
7.
Nat Commun ; 11(1): 5881, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33208748

ABSTRACT

Microbiota-host-diet interactions contribute to the development of metabolic diseases. Imidazole propionate is a novel microbially produced metabolite from histidine, which impairs glucose metabolism. Here, we show that subjects with prediabetes and diabetes in the MetaCardis cohort from three European countries have elevated serum imidazole propionate levels. Furthermore, imidazole propionate levels were increased in subjects with low bacterial gene richness and Bacteroides 2 enterotype, which have previously been associated with obesity. The Bacteroides 2 enterotype was also associated with increased abundance of the genes involved in imidazole propionate biosynthesis from dietary histidine. Since patients and controls did not differ in their histidine dietary intake, the elevated levels of imidazole propionate in type 2 diabetes likely reflects altered microbial metabolism of histidine, rather than histidine intake per se. Thus the microbiota may contribute to type 2 diabetes by generating imidazole propionate that can modulate host inflammation and metabolism.


Subject(s)
Diabetes Mellitus, Type 2/microbiology , Gastrointestinal Microbiome , Imidazoles/blood , Adult , Aged , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/metabolism , Cohort Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Female , Histidine/metabolism , Humans , Male , Middle Aged
8.
Mol Metab ; 39: 101007, 2020 09.
Article in English | MEDLINE | ID: mdl-32360426

ABSTRACT

OBJECTIVE: Obesity is characterized by systemic and low-grade tissue inflammation. In the intestine, alteration of the intestinal barrier and accumulation of inflammatory cells in the epithelium are important contributors of gut inflammation. Recent studies demonstrated the role of the aryl hydrocarbon receptor (AhR) in the maintenance of immune cells at mucosal barrier sites. A wide range of ligands of external and local origin can activate this receptor. We studied the causal relationship between AhR activation and gut inflammation in obesity. METHODS: Jejunum samples from subjects with normal weight and severe obesity were phenotyped according to T lymphocyte infiltration in the epithelium from lamina propria and assayed for the mRNA level of AhR target genes. The effect of an AhR agonist was studied in mice and Caco-2/TC7 cells. AhR target gene expression, permeability to small molecules and ions, and location of cell-cell junction proteins were recorded under conditions of altered intestinal permeability. RESULTS: We showed that a low AhR tone correlated with a high inflammatory score in the intestinal epithelium in severe human obesity. Moreover, AhR activation protected junctional complexes in the intestinal epithelium in mice challenged by an oral lipid load. AhR ligands prevented chemically induced damage to barrier integrity and cytokine expression in Caco-2/TC7 cells. The PKC and p38MAPK signaling pathways were involved in this AhR action. CONCLUSIONS: The results of these series of human, mouse, and cell culture experiments demonstrate the protective effect of AhR activation in the intestine targeting particularly tight junctions and cytokine expression. We propose that AhR constitutes a valuable target to protect intestinal functions in metabolic diseases, which can be achieved in the future via food or drug ligands.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Intestinal Mucosa/metabolism , Obesity/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Adiposity/genetics , Adult , Aged , Aged, 80 and over , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers , Cell Line , Comorbidity , Cytokines/metabolism , Epithelial Cells/metabolism , Female , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Jejunum/metabolism , Lipid Metabolism , MAP Kinase Signaling System , Male , Mice , Middle Aged , Models, Biological , Obesity/etiology , Obesity/pathology , Permeability , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism
9.
Acad Med ; 95(1): 151-156, 2020 01.
Article in English | MEDLINE | ID: mdl-31335813

ABSTRACT

PURPOSE: Using item analyses is an important quality-monitoring strategy for written exams. Authors urge caution as statistics may be unstable with small cohorts, making application of guidelines potentially detrimental. Given the small cohorts common in health professions education, this study's aim was to determine the impact of cohort size on outcomes arising from the application of item analysis guidelines. METHOD: The authors performed a Monte Carlo simulation study in fall 2015 to examine the impact of applying 2 commonly used item analysis guidelines on the proportion of items removed and overall exam reliability as a function of cohort size. Three variables were manipulated: Cohort size (6 levels), exam length (6 levels), and exam difficulty (3 levels). Study parameters were decided based on data provided by several Canadian medical schools. RESULTS: The analyses showed an increase in proportion of items removed with decreases in exam difficulty and decreases in cohort size. There was no effect of exam length on this outcome. Exam length had a greater impact on exam reliability than did cohort size after applying item analysis guidelines. That is, exam reliability decreased more with shorter exams than with smaller cohorts. CONCLUSIONS: Although program directors and assessment creators have little control over their cohort sizes, they can control the length of their exams. Creating longer exams makes it possible to remove items without as much negative impact on the exam's reliability relative to shorter exams, thereby reducing the negative impact of small cohorts when applying item removal guidelines.


Subject(s)
Curriculum/standards , Educational Measurement/standards , Health Occupations/education , Schools, Medical/statistics & numerical data , Canada/epidemiology , Cohort Studies , Educational Measurement/statistics & numerical data , Evaluation Studies as Topic , Guidelines as Topic , Health Occupations/standards , Humans , Monte Carlo Method , Psychometrics/methods , Reproducibility of Results , Time Factors
10.
J Allergy Clin Immunol ; 143(4): 1575-1585.e4, 2019 04.
Article in English | MEDLINE | ID: mdl-30554723

ABSTRACT

BACKGROUND: Commensals induce local IgA responses essential to the induction of tolerance to gut microbiota, but it remains unclear whether antimicrobiota responses remain confined to the gut. OBJECTIVE: The aim of this study was to investigate systemic and intestinal responses against the whole microbiota under homeostatic conditions and in the absence of IgA. METHODS: We analyzed blood and feces from healthy donors, patients with selective IgA deficiency (SIgAd), and patients with common variable immunodeficiency (CVID). Immunoglobulin-coated bacterial repertoires were analyzed by using combined bacterial fluorescence-activated cell sorting and 16S rRNA sequencing. Bacterial lysates were probed by using Western blot analysis with healthy donor sera. RESULTS: Although absent from the healthy gut, serum antimicrobiota IgG are present in healthy subjects and increased in patients with SIgAd. IgG converges with nonoverlapping secretory IgA specificities to target the same bacteria. Each individual subject targets a diverse microbiota repertoire with a proportion that correlates inversely with systemic inflammation. Finally, intravenous immunoglobulin preparations target CVID gut microbiota much less efficiently than healthy microbiota. CONCLUSION: Secretory IgA and systemic IgG converge to target gut microbiota at the cellular level. SIgAd-associated inflammation is inversely correlated with systemic anticommensal IgG responses, which might serve as a second line of defense. We speculate that patients with SIgAd could benefit from oral IgA supplementation. Our data also suggest that intravenous immunoglobulin preparations can be supplemented with IgG from IgA-deficient patient pools to offer better protection against gut bacterial translocations in patients with CVID.


Subject(s)
Gastrointestinal Microbiome/immunology , Immunoglobulin A, Secretory/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Antibodies, Bacterial/immunology , Common Variable Immunodeficiency/immunology , Feces/chemistry , Humans , IgA Deficiency/immunology
11.
Article in English | MEDLINE | ID: mdl-29936369

ABSTRACT

In this study, we describe the development of a SFC-MS method for the quality control of cannabis plants that could be potentially adulterated with synthetic cannabinoids. Considering the high number of already available synthetic cannabinoids and the high rate of development of novel structures, we aimed to develop a generic method suitable for the analysis of a large panel of substances using seventeen synthetic cannabinoids from multiple classes as model compounds. Firstly, a suitable column was chosen after a screening phase. Secondly, optimal operating conditions were obtained following a robust optimization strategy based on a design of experiments and design space methodology (DoE-DS). Finally, the quantitative performances of the method were assessed with a validation according to the total error approach. The developed method has a run time of 9.4 min. It uses a simple modifier composition of methanol with 2% H2O and requires minimal sample preparation. It can chromatographically separate natural cannabinoids (except THC-A and CBD-A) from the synthetics assessed. Also, the use of mass spectrometry provides sensitivity and specificity. Moreover, this quality by design (QbD) approach permits the tuning of the method (within the DS) during routine analysis to achieve a desirable separation since the future compounds that should be analyzed could be unknown. The method was validated for the quantitation of a selected synthetic cannabinoid in fiber-type cannabis matrix over the range of 2.5% - 7.5% (w/w) with LOD value as low as 14.4 ng/mL. This generic method should be easy to implement in customs or QC laboratories in the context of counterfeit drugs tracking.


Subject(s)
Cannabinoids/analysis , Chromatography, Supercritical Fluid/methods , Mass Spectrometry/methods , Medical Marijuana/analysis , Medical Marijuana/standards , Cannabinoids/chemistry , Cannabinoids/standards , Drug Contamination , Linear Models , Reproducibility of Results , Research Design , Sensitivity and Specificity
12.
FASEB J ; : fj201800052RR, 2018 Apr 27.
Article in English | MEDLINE | ID: mdl-29957059

ABSTRACT

The disruption of systemic immune homeostasis is a key mediator in the progression of cardiometabolic diseases (CMDs). We aimed to extend knowledge regarding the clinical relevance of CMD-associated variation of circulating mucosal-associated invariant T (MAIT) cell abundance and to explore underlying cellular mechanisms. We analyzed cross-sectional data from 439 participants of the Metagenomics in Cardiometabolic Diseases (MetaCardis) study, stratified into 6 groups: healthy control subjects and patients with metabolic syndrome (MS), obesity, type 2 diabetes mellitus (T2DM), and coronary artery disease (CAD) without, or with congestive heart failure (CAD-CHF). Blood MAIT cell frequency was significantly decreased in all CMD groups, including early (MS) and later (CAD and CAD-CHF) stages of disease progression. Reduced MAIT cell abundance was associated with increased glycosylated hemoglobin, inflammation markers, and deterioration of cardiac function. Glucose dose dependently promoted MAIT cell apoptosis in vitro, independently of anti-CD3 and cytokine-mediated activation. This outcome suggests the prominence of metabolic over an antigenic or cytokine-rich environment to promote MAIT cell reduction in patients with CMD. In summary, all stages of CMDs are characterized by reduced circulating MAIT cells. Chronically elevated blood glucose levels could contribute to this decline. These data extend the pathologic relevance of MAIT cell loss and suggest that MAIT cell abundance may serve as an indicator of cardiometabolic health.-Touch, S., Assmann, K. E., Aron-Wisnewsky, J., Marquet, F., Rouault, C., Fradet, M., Mosbah, H., MetaCardis Consortium, Isnard, R., Helft, G., Lehuen, A., Poitou, C., Clément, K., André, S. Mucosal-associated invariant T (MAIT) cells are depleted and prone to apoptosis in cardiometabolic disorders.

13.
Perspect Med Educ ; 7(2): 83-92, 2018 04.
Article in English | MEDLINE | ID: mdl-29294255

ABSTRACT

INTRODUCTION: With the Standards voicing concern for the appropriateness of response processes, we need to explore strategies that would allow us to identify inappropriate rater response processes. Although certain statistics can be used to help detect rater bias, their use is complicated by either a lack of data about their actual power to detect rater bias or the difficulty related to their application in the context of health professions education. This exploratory study aimed to establish the worthiness of pursuing the use of l z to detect rater bias. METHODS: We conducted a Monte Carlo simulation study to investigate the power of a specific detection statistic, that is: the standardized likelihood l z person-fit statistics (PFS). Our primary outcome was the detection rate of biased raters, namely: raters whom we manipulated into being either stringent (giving lower scores) or lenient (giving higher scores), using the l z statistic while controlling for the number of biased raters in a sample (6 levels) and the rate of bias per rater (6 levels). RESULTS: Overall, stringent raters (M = 0.84, SD = 0.23) were easier to detect than lenient raters (M = 0.31, SD = 0.28). More biased raters were easier to detect then less biased raters (60% bias: 62, SD = 0.37; 10% bias: 43, SD = 0.36). DISCUSSION: The PFS l z seems to offer an interesting potential to identify biased raters. We observed detection rates as high as 90% for stringent raters, for whom we manipulated more than half their checklist. Although we observed very interesting results, we cannot generalize these results to the use of PFS with estimated item/station parameters or real data. Such studies should be conducted to assess the feasibility of using PFS to identify rater bias.


Subject(s)
Bias , Educational Measurement/standards , Observer Variation , Research Personnel/psychology , Analysis of Variance , Humans , Monte Carlo Method , Research Personnel/standards
14.
Bioanalysis ; 10(2): 107-124, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29236519

ABSTRACT

During the last years, chemistry was involved in the worldwide effort toward environmental problems leading to the birth of green chemistry. In this context, green analytical tools were developed as modern Supercritical Fluid Chromatography in the field of separative techniques. This chromatographic technique knew resurgence a few years ago, thanks to its high efficiency, fastness and robustness of new generation equipment. These advantages and its easy hyphenation to MS fulfill the requirements of bioanalysis regarding separation capacity and high throughput. In the present paper, the technical aspects focused on bioanalysis specifications will be detailed followed by a critical review of bioanalytical supercritical fluid chromatography methods published in the literature.


Subject(s)
Biological Assay/methods , Chromatography, Supercritical Fluid/methods , Humans
15.
Curr Diab Rep ; 17(9): 81, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28779366

ABSTRACT

PURPOSE OF THE REVIEW: Obesity and type 2 diabetes (T2D) are considered chronic inflammatory diseases. While early publications have reported the implication of innate immune cells such as macrophages to promote systemic inflammation and metabolic dysfunctions, recent publications underline the alterations of the T cell compartment in human obesity and type 2 diabetes. These recent findings are the focus of this review. RECENT FINDINGS: In humans, obesity and T2D induce the expansion of proinflammatory T cells such as CD4 Th1, Th17, and CD8 populations, whereas innate T cells such as MAIT and iNKT cells are decreased. These alterations reflect a loss of total T cell homeostasis that may contribute to tissue and systemic inflammation. Whether these changes are adaptive to nutritional variations and/or contribute to the progression of metabolic diseases remains to be clarified. T cell phenotyping may improve obese and/or T2D patient stratification with therapeutic and prognostic implications.


Subject(s)
Diabetes Mellitus, Type 2/immunology , Obesity/immunology , T-Lymphocytes/immunology , Diabetes Mellitus, Type 2/complications , Humans , Immunity , Inflammation/immunology , Inflammation/pathology , Lymphocyte Subsets/immunology , Obesity/complications
16.
Sci Rep ; 7(1): 3000, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28592801

ABSTRACT

Adipose tissue contains a variety of immune cells, which vary in abundance and phenotype with obesity. The contribution of immune cell-derived factors to inflammatory, fibrotic and metabolic alterations in adipose tissue is not well established in human obesity. Human primary adipose tissue cells, including pre-adipocytes, endothelial cells and mature adipocytes, were used to investigate deregulation of cell- and pathway-specific gene profiles. Among factors known to alter adipose tissue biology, we focus on inflammatory (IL-1ß and IL-17) and pro-fibrotic (TGF-ß1) factors. rIL-1ß and rIL-17 induced concordant pro-inflammatory transcriptional programs in pre-adipocytes and endothelial cells, with a markedly more potent effect of IL-1ß than IL-17. None of these cytokines had significant effect on fibrogenesis-related gene expression, contrasting with rTGF-ß1-induced up-regulation of extracellular matrix components and pro-fibrotic factors. In mature adipocytes, all three factors promoted down-regulation of genes functionally involved in lipid storage and release. IL-1ß and IL-17 impacted adipocyte metabolic genes in relation with their respective pro-inflammatory capacity, while the effect of TGF-ß1 occurred in face of an anti-inflammatory signature. These data revealed that IL-1ß and IL-17 had virtually no effect on pro-fibrotic alterations but promote inflammation and metabolic dysfunction in human adipose tissue, with a prominent role for IL-1ß.


Subject(s)
Adipose Tissue/pathology , Cytokines/metabolism , Inflammation/pathology , Obesity/pathology , Adipocytes/pathology , Cells, Cultured , Endothelial Cells/pathology , Female , Gene Expression Profiling , Humans , Male , Middle Aged
17.
FASEB J ; 30(1): 241-51, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26362817

ABSTRACT

In mice, nutritional supplementation with the trans-10,cis-12 isomer of linoleic acid (t10,c12-CLA) promotes lipoatrophy, hyperinsulinemia, and macrophage infiltration in white adipose tissue (WAT). We explored the dynamics of these interrelated responses over 2 consecutive 7 d periods of t10,c12-CLA administration and withdrawal. t10,c12-CLA down-regulated lipogenic and lipolytic gene expression and increased collagen deposition, but with no evidence of cross-linking. An abundant CD45(+) cell infiltrate, comprising prominently CD206(+)CD11c(-) macrophages, was found in WAT in association with an anti-inflammatory gene signature. Infiltration of natural killer (NK) and dendritic cells contributed to WAT's innate immune response to t10,c12-CLA. Less abundant adaptive immune cells colonized WAT, including B, NK T, γδ T, and αß T cells. By contrast, T-regulatory cell abundance was not affected. Interruption of treatment allowed recovery of WAT mass and normalization of insulinemia, coincident with regain of WAT homeostasis owing to a coordinated reversion of genic, structural, and immune deregulations. These data revealed a striking resilience of WAT after a short-term metabolic injury induced by t10,c12-CLA, which relies on alternatively activated M2 macrophage engagement. In addition, the temporal links between variations in WAT alterations and insulinemia upon t10,c12-CLA manipulation strengthen the view that WAT dysfunctional status is critically involved in altered glucose homeostasis.


Subject(s)
Adipose Tissue, White/drug effects , Linoleic Acids, Conjugated/pharmacology , Macrophage Activation , Macrophages/drug effects , Adaptation, Physiological , Adipose Tissue, White/cytology , Animals , Cells, Cultured , Female , Macrophages/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes/drug effects
18.
Endocrinology ; 156(11): 4047-58, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26295369

ABSTRACT

Prior studies demonstrated increased plasma IgE in diabetic patients, but the direct participation of IgE in diabetes or obesity remains unknown. This study found that plasma IgE levels correlated inversely with body weight, body mass index, and body fat mass among a population of randomly selected obese women. IgE receptor FcϵR1-deficient (Fcer1a(-/-)) mice and diet-induced obesity (DIO) mice demonstrated that FcϵR1 deficiency in DIO mice increased food intake, reduced energy expenditure, and increased body weight gain but improved glucose tolerance and glucose-induced insulin secretion. White adipose tissue from Fcer1a(-/-) mice showed an increased expression of phospho-AKT, CCAAT/enhancer binding protein-α, peroxisome proliferator-activated receptor-γ, glucose transporter-4 (Glut4), and B-cell lymphoma 2 (Bcl2) but reduced uncoupling protein 1 (UCP1) and phosphorylated c-Jun N-terminal kinase (JNK) expression, tissue macrophage accumulation, and apoptosis, suggesting that IgE reduces adipogenesis and glucose uptake but induces energy expenditure, adipocyte apoptosis, and white adipose tissue inflammation. In 3T3-L1 cells, IgE inhibited the expression of CCAAT/enhancer binding protein-α and peroxisome proliferator-activated receptor-γ, and preadipocyte adipogenesis and induced adipocyte apoptosis. IgE reduced the 3T3-L1 cell expression of Glut4, phospho-AKT, and glucose uptake, which concurred with improved glucose tolerance in Fcer1a(-/-) mice. This study established two novel pathways of IgE in reducing body weight gain in DIO mice by suppressing adipogenesis and inducing adipocyte apoptosis while worsening glucose tolerance by reducing Glut4 expression, glucose uptake, and insulin secretion.


Subject(s)
Energy Metabolism/genetics , Obesity/genetics , Receptors, IgE/genetics , Weight Gain/genetics , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Diet, High-Fat/adverse effects , Female , Gene Expression , Glucose Tolerance Test , Humans , Immunoblotting , Immunoglobulin E/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity/etiology , Obesity/metabolism , Obesity, Morbid/blood , PPAR gamma/genetics , PPAR gamma/metabolism , RNA Interference , Receptors, IgE/deficiency , Reverse Transcriptase Polymerase Chain Reaction
19.
Cell Metab ; 22(1): 113-24, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26094890

ABSTRACT

In obesity, insulin resistance is linked to inflammation in several tissues. Although the gut is a very large lymphoid tissue, inflammation in the absorptive small intestine, the jejunum, where insulin regulates lipid and sugar absorption is unknown. We analyzed jejunal samples of 185 obese subjects stratified in three metabolic groups: without comorbidity, suffering from obesity-related comorbidity, and diabetic, versus 33 lean controls. Obesity increased both mucosa surface due to lower cell apoptosis and innate and adaptive immune cell populations. The preferential CD8αß T cell location in epithelium over lamina propria appears a hallmark of obesity. Cytokine secretion by T cells from obese, but not lean, subjects blunted insulin signaling in enterocytes relevant to apical GLUT2 mislocation. Statistical links between T cell densities and BMI, NAFLD, or lipid metabolism suggest tissue crosstalk. Obesity triggers T-cell-mediated inflammation and enterocyte insulin resistance in the jejunum with potential broader systemic implications.


Subject(s)
Enterocytes/pathology , Inflammation/complications , Insulin/immunology , Jejunum/pathology , Obesity/complications , T-Lymphocytes/pathology , Adult , CD8 Antigens/immunology , Cells, Cultured , Enterocytes/immunology , Female , Glucose Transporter Type 2/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Insulin Resistance , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Jejunum/cytology , Jejunum/immunology , Male , Middle Aged , Obesity/immunology , Obesity/pathology , Signal Transduction , T-Lymphocytes/immunology
20.
Nat Med ; 21(6): 610-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25939064

ABSTRACT

Accumulation of visceral adipose tissue correlates with elevated inflammation and increased risk of metabolic diseases. However, little is known about the molecular mechanisms that control its pathological expansion. Transcription factor interferon regulatory factor 5 (IRF5) has been implicated in polarizing macrophages towards an inflammatory phenotype. Here we demonstrate that mice lacking Irf5, when placed on a high-fat diet, show no difference in the growth of their epididymal white adipose tissue (epiWAT) but they show expansion of their subcutaneous white adipose tissue, as compared to wild-type (WT) mice on the same diet. EpiWAT from Irf5-deficient mice is marked by accumulation of alternatively activated macrophages, higher collagen deposition that restricts adipocyte size, and enhanced insulin sensitivity compared to epiWAT from WT mice. In obese individuals, IRF5 expression is negatively associated with insulin sensitivity and collagen deposition in visceral adipose tissue. Genome-wide analysis of gene expression in adipose tissue macrophages highlights the transforming growth factor ß1 (TGFB1) gene itself as a direct target of IRF5-mediated inhibition. This study uncovers a new function for IRF5 in controlling the relative mass of different adipose tissue depots and thus insulin sensitivity in obesity, and it suggests that inhibition of IRF5 may promote a healthy metabolic state during this condition.


Subject(s)
Adipose Tissue, White/metabolism , Inflammation/genetics , Interferon Regulatory Factors/genetics , Obesity/genetics , Animals , Diet, High-Fat , Gene Expression Regulation , Humans , Inflammation/drug therapy , Inflammation/pathology , Insulin Resistance/genetics , Macrophages , Mice , Obesity/drug therapy , Obesity/pathology , Transforming Growth Factor beta1/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL