Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 16(4): 513-526, 2023 08.
Article in English | MEDLINE | ID: mdl-37302712

ABSTRACT

Obesity and type 2 diabetes (T2D) have been found to be associated with abnormalities in several organs, including the intestine. These conditions can lead to changes in gut homeostasis, compromising tolerance to luminal antigens and increasing susceptibility to food allergies. The underlying mechanisms for this phenomenon are not yet fully understood. In this study, we investigated changes in the intestinal mucosa of diet-induced obese mice and found that they exhibited increased gut permeability and reduced Treg cells frequency. Upon oral treatment with ovalbumin (OVA), obese mice failed to develop oral tolerance. However, hyperglycemia treatment improved intestinal permeability and oral tolerance induction in mice. Furthermore, we observed that obese mice exhibited a more severe food allergy to OVA, and this allergy was alleviated after treatment with a hypoglycemic drug. Importantly, our findings were translated to obese humans. Individuals with T2D had higher serum IgE levels and downregulated genes related to gut homeostasis. Taken together, our results suggest that obesity-induced hyperglycemia can lead to a failure in oral tolerance and to exacerbation of food allergy. These findings shed light on the mechanisms underlying the relationship among obesity, T2D, and gut mucosal immunity, which could inform the development of new therapeutic approaches.


Subject(s)
Diabetes Mellitus, Type 2 , Food Hypersensitivity , Humans , Mice , Animals , Mice, Obese , Obesity , Immune Tolerance , Allergens , Administration, Oral , Ovalbumin , Mice, Inbred BALB C
2.
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
3.
Quant Imaging Med Surg ; 11(5): 2162-2168, 2021 May.
Article in English | MEDLINE | ID: mdl-33936996

ABSTRACT

To determine whether magnetic resonance imaging (MRI) when used in an optimal ex vivo setting can help detecting and quantifying the 3D fibrosis fraction in human subcutaneous adipose tissue (SAT) samples, as compared to histology. This prospective observational study was approved by our institutional review board 3D MRI acquisitions were performed at 4.0 T (Bruker) on XX human SAT samples (around 1 cm3) collected from biopsy in morbidly obese patients. Such acquisitions included saturation-recovery T1 mapping (spatial resolution: 200 µm, acquisition time: ~16 minutes) and DIXON imaging (spatial resolution: 200 µm, acquisition time: ~20 minutes). After MRI, histological quantification of fibrosis was performed using picrosirius staining. T1 maps were clustered based on a k-means algorithm allowing quantification of fibrosis within the adipose tissue and percentage of fibrosis over the entire sample volume was calculated. Fat maps were computed from DIXON in-phase and out-of-phase images. The 3D MRI fibrosis percentage within the SAT samples were comprised between 6% and 15%. Excellent correlations and levels of agreement were observed between single slice MRI and histology (r=0.9, P=0.08) and between 3D MRI and histology in terms fibrosis percentages within SAT samples (r=0.9, P=0.01). High Field ex vivo MRI was able to quantify fibrosis in human SAT samples with high agreement with histology and moreover to provide 3D SAT fibrosis quantification avoiding histological sampling errors.

4.
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
6.
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
7.
EBioMedicine ; 58: 102895, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32739864

ABSTRACT

BACKGROUND: Bariatric surgery is an effective treatment for type 2 diabetes. Early post-surgical enhancement of insulin secretion is key for diabetes remission. The full complement of mechanisms responsible for improved pancreatic beta cell functionality after bariatric surgery is still unclear. Our aim was to identify pathways, evident in the islet transcriptome, that characterize the adaptive response to bariatric surgery independently of body weight changes. METHODS: We performed entero-gastro-anastomosis (EGA) with pyloric ligature in leptin-deficient ob/ob mice as a surrogate of Roux-en-Y gastric bypass (RYGB) in humans. Multiple approaches such as determination of glucose tolerance, GLP-1 and insulin secretion, whole body insulin sensitivity, ex vivo glucose-stimulated insulin secretion (GSIS) and functional multicellular Ca2+-imaging, profiling of mRNA and of miRNA expression were utilized to identify significant biological processes involved in pancreatic islet recovery. FINDINGS: EGA resolved diabetes, increased pancreatic insulin content and GSIS despite a persistent increase in fat mass, systemic and intra-islet inflammation, and lipotoxicity. Surgery differentially regulated 193 genes in the islet, most of which were involved in the regulation of glucose metabolism, insulin secretion, calcium signaling or beta cell viability, and these were normalized alongside changes in glucose metabolism, intracellular Ca2+ dynamics and the threshold for GSIS. Furthermore, 27 islet miRNAs were differentially regulated, four of them hubs in a miRNA-gene interaction network and four others part of a blood signature of diabetes resolution in ob/ob mice and in humans. INTERPRETATION: Taken together, our data highlight novel miRNA-gene interactions in the pancreatic islet during the resolution of diabetes after bariatric surgery that form part of a blood signature of diabetes reversal. FUNDING: European Union's Horizon 2020 research and innovation programme via the Innovative Medicines Initiative 2 Joint Undertaking (RHAPSODY), INSERM, Société Francophone du Diabète, Institut Benjamin Delessert, Wellcome Trust Investigator Award (212625/Z/18/Z), MRC Programme grants (MR/R022259/1, MR/J0003042/1, MR/L020149/1), Diabetes UK (BDA/11/0004210, BDA/15/0005275, BDA 16/0005485) project grants, National Science Foundation (310030-188447), Fondation de l'Avenir.


Subject(s)
Diabetes Mellitus, Type 2/surgery , Gene Regulatory Networks , Insulin-Secreting Cells/chemistry , MicroRNAs/genetics , Obesity/surgery , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Gastric Bypass , Gene Expression Profiling , Gene Expression Regulation , Glucagon-Like Peptide 1/metabolism , Glucose Tolerance Test , Humans , Insulin/metabolism , Male , Mice , Mice, Obese , Obesity/genetics , Obesity/metabolism
8.
FASEB J ; 33(4): 4741-4754, 2019 04.
Article in English | MEDLINE | ID: mdl-30608881

ABSTRACT

Lipidomic techniques can improve our understanding of complex lipid interactions that regulate metabolic diseases. Here, a serum phospholipidomics analysis identified associations between phosphatidylglycerols (PGs) and gut microbiota dysbiosis. Compared with the other phospholipids, serum PGs were the most elevated in patients with low microbiota gene richness, which were normalized after a dietary intervention that restored gut microbial diversity. Serum PG levels were positively correlated with metagenomic functional capacities for bacterial LPS synthesis and host markers of low-grade inflammation; transcriptome databases identified PG synthase, the first committed enzyme in PG synthesis, as a potential mediator. Experiments in mice and cultured human-derived macrophages demonstrated that LPS induces PG release. Acute PG treatment in mice altered adipose tissue gene expression toward remodeling and inhibited ex vivo lipolysis in adipose tissue, suggesting that PGs favor lipid storage. Indeed, several PG species were associated with the severity of obesity in mice and humans. Finally, despite enrichment in PGs in bacterial membranes, experiments employing gnotobiotic mice colonized with recombinant PG overproducing Lactococcus lactis showed limited direct contribution of microbial PGs to the host. In summary, PGs are inflammation-responsive lipids indirectly regulated by the gut microbiota via endotoxins and regulate adipose tissue homeostasis in obesity.-Kayser, B. D., Lhomme, M., Prifti, E., Da Cunha, C., Marquet, F., Chain, F., Naas, I., Pelloux, V., Dao, M.-C., Kontush, A., Rizkalla, S. W., Aron-Wisnewsky, J., Bermúdez-Humarán, L. G., Oakley, F., Langella, P., Clément, K., Dugail, I. Phosphatidylglycerols are induced by gut dysbiosis and inflammation, and favorably modulate adipose tissue remodeling in obesity.


Subject(s)
Adipose Tissue/metabolism , Dysbiosis/metabolism , Inflammation/metabolism , Obesity/metabolism , Phosphatidylglycerols/metabolism , Animals , Female , Humans , Lipidomics/methods , Lipolysis/physiology , Male , Metagenomics/methods , Mice
9.
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
10.
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.

11.
Nat Commun ; 9(1): 725, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29463843

ABSTRACT

In systemic lupus erythematosus (SLE), autoantibody production can lead to kidney damage and failure, known as lupus nephritis. Basophils amplify the synthesis of autoantibodies by accumulating in secondary lymphoid organs. Here, we show a role for prostaglandin D2 (PGD2) in the pathophysiology of SLE. Patients with SLE have increased expression of PGD2 receptors (PTGDR) on blood basophils and increased concentration of PGD2 metabolites in plasma. Through an autocrine mechanism dependent on both PTGDRs, PGD2 induces the externalization of CXCR4 on basophils, both in humans and mice, driving accumulation in secondary lymphoid organs. Although PGD2 can accelerate basophil-dependent disease, antagonizing PTGDRs in mice reduces lupus-like disease in spontaneous and induced mouse models. Our study identifies the PGD2/PTGDR axis as a ready-to-use therapeutic modality in SLE.


Subject(s)
Basophils/immunology , Lupus Erythematosus, Systemic/immunology , Lymphatic System/immunology , Prostaglandin D2/immunology , Adult , Animals , Female , Humans , Lupus Erythematosus, Systemic/blood , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Prostaglandin D2/blood , Receptors, CXCR4/blood , Receptors, CXCR4/immunology , Receptors, Immunologic/blood , Receptors, Immunologic/immunology , Receptors, Prostaglandin/blood , Receptors, Prostaglandin/immunology , Signal Transduction/immunology , Young Adult
12.
Front Microbiol ; 9: 3289, 2018.
Article in English | MEDLINE | ID: mdl-30692975

ABSTRACT

The intestinal microbiota and its functions are intricately interwoven with host physiology. Colonizing rodents with donor microbiota provides insights into host-microbiota interactions characterization and the understanding of disease physiopathology. However, a better assessment of inoculation methods and recipient mouse models is needed. Here, we compare the engraftment at short and long term of genetically obese mice microbiota in germ-free (GF) mice and juvenile and adult specific pathogen free (SPF) mice. We also tested the effects of initial microbiota depletion before microbiota transfer. In the present work, donor microbiota engraftment was better in juvenile SPF mice than in adult SPF mice. In juvenile mice, initial microbiota depletion using laxatives or antibiotics improved donor microbiota engraftment 9 weeks but not 3 weeks after microbiota transfer. Microbiota-depleted juvenile mice performed better than GF mice 3 weeks after the microbiota transfer. However, 9 weeks after transfer, colonized GF mice microbiota had the lowest Unifrac distance to the donor microbiota. Colonized GF mice were also characterized by a chronic alteration in intestinal absorptive function. With these collective results, we show that the use of juvenile mice subjected to initial microbiota depletion constitutes a valid alternative to GF mice in microbiota transfer studies.

13.
Mol Immunol ; 63(1): 86-93, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24793464

ABSTRACT

Mast cells are hematopoietic cells involved in inflammation and immunity and have been recognized also as important effector cells in kidney inflammation. In humans, only a few mast cells reside in kidneys constitutively but in progressive renal diseases their numbers increase substantially representing an essential part of the interstitial infiltrate of inflammatory cells. Recent data obtained in experimental animal models have emphasized a complex role of these cells and the mediators they release as they have been shown both to promote, but also to protect from disease and fibrosis development. Sometimes conflicting results have been reported in similar models suggesting a very narrow window between these activities depending on the pathophysiological context. Interestingly in mice, mast cell or mast cell mediator specific actions became also apparent in the absence of significant mast cell kidney infiltration supporting systemic or regional actions via draining lymph nodes or kidney capsules. Many of their activities rely on the capacity of mast cells to release, in a timely controlled manner, a wide range of inflammatory mediators, which can promote anti-inflammatory actions and repair activities that contribute to healing, but in some circumstances or in case of inappropriate regulation may also promote kidney disease.


Subject(s)
Kidney/immunology , Kidney/pathology , Mast Cells/immunology , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/pathology , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Animals , Disease Models, Animal , Fibrosis , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Humans , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Mice
14.
J Immunol ; 193(12): 5883-93, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25385823

ABSTRACT

Swine skin is one of the best structural models for human skin, widely used to probe drug transcutaneous passage and to test new skin vaccination devices. However, little is known about its composition in immune cells, and among them dendritic cells (DC), that are essential in the initiation of the immune response. After a first seminal work describing four different DC subpopulations in pig skin, we hereafter deepen the characterization of these cells, showing the similarities between swine DC subsets and their human counterparts. Using comparative transcriptomic study, classical phenotyping as well as in vivo and in vitro functional studies, we show that swine CD163(pos) dermal DC (DDC) are transcriptomically similar to the human CD14(pos) DDC. CD163(pos) DDC are recruited in inflamed skin, they migrate in inflamed lymph but they are not attracted toward CCL21, and they modestly activate allogeneic CD8 T cells. We also show that CD163(low) DDC are transcriptomically similar to the human CD1a(pos) DDC. CD163(low) DDC migrate toward CCL21, they activate allogeneic CD8 and CD4 T cells and, like their potential human lung counterpart, they skew CD4 T cells toward a Th17 profile. We thus conclude that swine skin is a relevant model for human skin vaccination.


Subject(s)
Chemotaxis/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Langerhans Cells/immunology , Langerhans Cells/metabolism , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , Transcriptome , Animals , Antigens, CD1/genetics , Antigens, CD1/metabolism , Antigens, Surface/metabolism , Chemotaxis/genetics , Cytokines/biosynthesis , Gene Expression Profiling , Humans , Immunophenotyping , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Phenotype , Skin/immunology , Swine
15.
Vet Immunol Immunopathol ; 144(3-4): 430-6, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22015056

ABSTRACT

Dendritic cells (DC) in peripheral tissues are considered as immature cells that mature and migrate towards lymph nodes upon stimulation with pathogens. This commonly accepted paradigm is challenged by the fact that tolerance to peripheral self antigen is controlled by mature DC and that DC collected from afferent lymph draining different tissues from several species, in the absence of pathogen signaling, were inconsistently found to be either at a mature or semi-mature state. In order to better define the maturation state of DC that migrate in lymph in absence of pathogen stimulation, we compared skin lymph DC to resident and LPS (lipopolysaccharide)-activated skin DC thanks to the establishment of a mini-pig model of lymph duct cannulation. Based on their co-stimulatory molecules expression and endocytotic capacities, pig lymph skin DC were found at an intermediate state of maturation between resident and LPS-activated skin DC and were fully capable of allogeneic T cell stimulation. Furthermore, lymph skin DC could be further matured by LPS or influenza stimulation. Thus, using the pig skin model which is relevant to human, we show that skin-derived DC constantly migrate at an intermediate state of maturation that can be further enhanced upon appropriate stimulation.


Subject(s)
Langerhans Cells/physiology , Swine/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/physiology , Cell Movement/immunology , Cell Movement/physiology , Langerhans Cells/immunology , Phenotype , Skin/cytology , Skin/immunology
16.
PLoS One ; 6(1): e16320, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21298011

ABSTRACT

Transcutaneous delivery of vaccines to specific skin dendritic cells (DC) subsets is foreseen as a promising strategy to induce strong and specific types of immune responses such as tolerance, cytotoxicity or humoral immunity. Because of striking histological similarities between human and pig skin, pig is recognized as the most suitable model to study the cutaneous delivery of medicine. Therefore improving the knowledge on swine skin DC subsets would be highly valuable to the skin vaccine field. In this study, we showed that pig skin DC comprise the classical epidermal langerhans cells (LC) and dermal DC (DDC) that could be divided in 3 subsets according to their phenotypes: (1) the CD163(neg)/CD172a(neg), (2) the CD163(high)CD172a(pos) and (3) the CD163(low)CD172a(pos) DDC. These subtypes have the capacity to migrate from skin to lymph node since we detected them in pseudo-afferent lymph. Extensive phenotyping with a set of markers suggested that the CD163(high) DDC resemble the antibody response-inducing human skin DC/macrophages whereas the CD163(neg)CD172(low) DDC share properties with the CD8(+) T cell response-inducing murine skin CD103(pos) DC. This work, by showing similarities between human, mouse and swine skin DC, establishes pig as a model of choice for the development of transcutaneous immunisation strategies targeting DC.


Subject(s)
Dendritic Cells/immunology , Langerhans Cells/cytology , Models, Animal , Administration, Cutaneous , Animals , Antigens, CD/analysis , Dendritic Cells/cytology , Humans , Immunophenotyping , Langerhans Cells/immunology , Lymph Nodes/cytology , Mice , Skin/cytology , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...