Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Elife ; 62017 06 28.
Article in English | MEDLINE | ID: mdl-28656887

ABSTRACT

Activation and increased numbers of inflammatory macrophages, in adipose tissue (AT) are deleterious in metabolic diseases. Up to now, AT macrophages (ATM) accumulation was considered to be due to blood infiltration or local proliferation, although the presence of resident hematopoietic stem/progenitor cells (Lin-/Sca+/c-Kit+; LSK phenotype) in the AT (AT-LSK) has been reported. By using transplantation of sorted AT-LSK and gain and loss of function studies we show that some of the inflammatory ATM inducing metabolic disease, originate from resident AT-LSK. Transplantation of AT-LSK sorted from high fat diet-fed (HFD) mice is sufficient to induce ATM accumulation, and to transfer metabolic disease in control mice. Conversely, the transplantation of control AT-LSK improves both AT-inflammation and glucose homeostasis in HFD mice. Our results clearly demonstrate that resident AT-LSK are one of the key point of metabolic disease, and could thus constitute a new promising therapeutic target to fight against metabolic disease.


Subject(s)
Adipose Tissue/physiology , Cell Proliferation , Diet/adverse effects , Metabolic Diseases , Myelopoiesis , Stem Cells/physiology , Animals , Macrophages/physiology , Mice
2.
J Exp Med ; 213(7): 1353-74, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27353089

ABSTRACT

Acute myocardial infarction (MI) is a severe ischemic disease responsible for heart failure and sudden death. Inflammatory cells orchestrate postischemic cardiac remodeling after MI. Studies using mice with defective mast/stem cell growth factor receptor c-Kit have suggested key roles for mast cells (MCs) in postischemic cardiac remodeling. Because c-Kit mutations affect multiple cell types of both immune and nonimmune origin, we addressed the impact of MCs on cardiac function after MI, using the c-Kit-independent MC-deficient (Cpa3(Cre/+)) mice. In response to MI, MC progenitors originated primarily from white adipose tissue, infiltrated the heart, and differentiated into mature MCs. MC deficiency led to reduced postischemic cardiac function and depressed cardiomyocyte contractility caused by myofilament Ca(2+) desensitization. This effect correlated with increased protein kinase A (PKA) activity and hyperphosphorylation of its targets, troponin I and myosin-binding protein C. MC-specific tryptase was identified to regulate PKA activity in cardiomyocytes via protease-activated receptor 2 proteolysis. This work reveals a novel function for cardiac MCs modulating cardiomyocyte contractility via alteration of PKA-regulated force-Ca(2+) interactions in response to MI. Identification of this MC-cardiomyocyte cross-talk provides new insights on the cellular and molecular mechanisms regulating the cardiac contractile machinery and a novel platform for therapeutically addressable regulators.


Subject(s)
Calcium Signaling , Calcium/metabolism , Mast Cells/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Myofibrils/metabolism , Animals , Carboxypeptidases A/genetics , Carboxypeptidases A/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Mice , Mice, Knockout , Myocardial Contraction/genetics , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Myofibrils/pathology , Proteolysis , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Receptor, PAR-2/genetics , Receptor, PAR-2/metabolism
3.
J Cell Physiol ; 230(12): 3076-83, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25989607

ABSTRACT

White adipose tissue (WAT) can be found in different locations in the body, and these different adipose deposits exhibit specific physiopathological importance according to the subcutaneous or abdominal locations. We have shown previously the presence of functional hematopoietic stem/progenitor cells (HSPC) in subcutaneous adipose tissue (SCAT). These cells exhibit a specific hematopoietic activity that contributes to the renewal of the immune cell compartment within this adipose deposit. In this study, we investigated whether HSPC can be found in visceral adipose tissue (VAT) and whether a putative difference in in situ hematopoiesis may be related to anatomical location and to site-specific immune cell content in VAT compared to SCAT. Therein, we identified for the first time the presence of HSPC in VAT. Using both in vitro assays and in vivo competitive repopulation experiments with sorted HSPC from VAT or SCAT, we showed that the hematopoietic activity of HSPC was lower in VAT, compared to SCAT. In addition, this altered hematopoietic activity of HSPC in VAT was due to their microenvironment, and may be related to a specific combination of secreted factors and extracellular matrix molecules expressed by adipose derived stromal cells. Our results indicate that WAT specific hematopoietic activity may be generalized to all adipose deposits, although with specificity according to the fat pad location. Considering the abundance of WAT in the body, this emphasizes the potential importance of this hematopoietic activity in physiopathological situations.


Subject(s)
Hematopoiesis, Extramedullary , Hematopoietic Stem Cells/physiology , Intra-Abdominal Fat/physiology , Subcutaneous Fat/physiology , Animals , Cell Communication , Cell Lineage , Cell Proliferation , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/immunology , Male , Mice, Inbred C57BL , Phenotype , Stem Cell Niche , Subcutaneous Fat/cytology , Subcutaneous Fat/immunology
4.
EMBO Mol Med ; 7(3): 259-74, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25666722

ABSTRACT

Pattern recognition receptors link metabolite and bacteria-derived inflammation to insulin resistance during obesity. We demonstrate that NOD2 detection of bacterial cell wall peptidoglycan (PGN) regulates metabolic inflammation and insulin sensitivity. An obesity-promoting high-fat diet (HFD) increased NOD2 in hepatocytes and adipocytes, and NOD2(-/-) mice have increased adipose tissue and liver inflammation and exacerbated insulin resistance during a HFD. This effect is independent of altered adiposity or NOD2 in hematopoietic-derived immune cells. Instead, increased metabolic inflammation and insulin resistance in NOD2(-/-) mice is associated with increased commensal bacterial translocation from the gut into adipose tissue and liver. An intact PGN-NOD2 sensing system regulated gut mucosal bacterial colonization and a metabolic tissue dysbiosis that is a potential trigger for increased metabolic inflammation and insulin resistance. Gut dysbiosis in HFD-fed NOD2(-/-) mice is an independent and transmissible factor that contributes to metabolic inflammation and insulin resistance when transferred to WT, germ-free mice. These findings warrant scrutiny of bacterial component detection, dysbiosis, and protective immune responses in the links between inflammatory gut and metabolic diseases, including diabetes.


Subject(s)
Bacteria/immunology , Diet/methods , Dysbiosis , Inflammation/pathology , Insulin Resistance , Nod2 Signaling Adaptor Protein/metabolism , Peptidoglycan/metabolism , Animals , Cell Wall/chemistry , Mice , Mice, Knockout , Peptidoglycan/analysis
5.
Mol Metab ; 2(3): 281-91, 2013.
Article in English | MEDLINE | ID: mdl-24049740

ABSTRACT

Metabolic endotoxemia triggers inflammation, targets cells from the stroma-vascular fraction of adipose depots, and metabolic disease. To identify these cells we here infused mice with lipopolysaccharides and showed by FACS analyses and BrdU staining that the number of small subcutaneous adipocytes, preadipocytes and macrophages increased in wild type but not in CD14-knockout (KO) mice. This mechanism was direct since in CD14KO mice grafted subcutaneously and simultaneously with fat pads from CD14KO and wild-type mice the concentration of cytokine mRNA was increased in the wild-type fat pad only. Conversely, the mRNA concentration of genes involved in glucose and lipid metabolism and the number of large adipocytes was reduced. Eventually, a pretreatment with LPS enhanced HFD-induced metabolic diseases. Altogether, these results show that metabolic endotoxemia increases the proliferation of preadipocytes through a CD14-dependent mechanism directly, without recruiting CD14-positive cells from non-adipose depot origin. This mechanism could precede the onset of metabolic diseases.

6.
Diabetologia ; 56(11): 2524-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23963324

ABSTRACT

AIMS/HYPOTHESIS: Circulating lipopolysaccharide-binding protein (LBP) is an acute-phase reactant known to be increased in obesity. We hypothesised that LBP is produced by adipose tissue (AT) in association with obesity. METHODS: LBP mRNA and LBP protein levels were analysed in AT from three cross-sectional (n = 210, n = 144 and n = 28) and three longitudinal (n = 8, n = 25, n = 20) human cohorts; in AT from genetically manipulated mice; in isolated adipocytes; and in human and murine cell lines. The effects of a high-fat diet and exposure to lipopolysaccharide (LPS) and peroxisome proliferator-activated receptor (PPAR)γ agonist were explored. Functional in vitro and ex vivo experiments were also performed. RESULTS: LBP synthesis and release was demonstrated to increase with adipocyte differentiation in human and mouse AT, isolated adipocytes and human and mouse cell lines (Simpson-Golabi-Behmel syndrome [SGBS], human multipotent adipose-derived stem [hMAD] and 3T3-L1 cells). AT LBP expression was robustly associated with inflammatory markers and increased with metabolic deterioration and insulin resistance in two independent cross-sectional human cohorts. AT LBP also increased longitudinally with weight gain and excessive fat accretion in both humans and mice, and decreased with weight loss (in two other independent cohorts), in humans with acquired lipodystrophy, and after ex vivo exposure to PPARγ agonist. Inflammatory agents such as LPS and TNF-α led to increased AT LBP expression in vivo in mice and in vitro, while this effect was prevented in Cd14-knockout mice. Functionally, LBP knockdown using short hairpin (sh)RNA or anti-LBP antibody led to increases in markers of adipogenesis and decreased adipocyte inflammation in human adipocytes. CONCLUSIONS/INTERPRETATION: Collectively, these findings suggest that LBP might have an essential role in inflammation- and obesity-associated AT dysfunction.


Subject(s)
Acute-Phase Proteins/metabolism , Adipocytes/metabolism , Adipose Tissue/pathology , Carrier Proteins/metabolism , Inflammation/metabolism , Membrane Glycoproteins/metabolism , Obesity/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adult , Animals , Humans , In Vitro Techniques , Insulin Resistance/physiology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Rosiglitazone , Thiazolidinediones/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
7.
J Nutr Biochem ; 24(7): 1266-75, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23333090

ABSTRACT

Lactoferrin is considered an epithelial protein present in different gland secretions. Administration of exogenous lactoferrin is also known to modulate adipogenesis and insulin action in human adipocytes. Here, we aimed to investigate lactoferrin gene expression (real-time polymerase chain reaction) and protein (enzyme-linked immunosorbent assay) levels in human (n=143) and mice adipose tissue samples, in adipose tissue fractions and during human preadipocyte and 3T3-L1 cell line differentiation, evaluating the effects of inducers (rosiglitazone) and disruptors (inflammatory factors) of adipocyte differentiation. Lactoferrin (LTF) gene and protein were detectable at relatively high levels in whole adipose tissue and isolated adipocytes in direct association with low-density lipoprotein-related protein 1 (LRP1, its putative receptor). Obese subjects with type 2 diabetes and increased triglycerides had the lowest levels of LTF gene expression in subcutaneous adipose tissue. Specifically, LTF gene expression was significantly increased in adipocytes, mainly from lean subjects, increasing during differentiation in parallel to adipogenic genes and gene markers of lipid droplets. The induction or disruption of adipogenesis led to concomitant changes (increase and decrease, respectively) of lactoferrin levels during adipocyte differentiation also in parallel to gene markers of adipogenesis and lipid droplet development. The administration of lactoferrin led to autopotentiated increased expression of the LTF gene. The decreased lactoferrin mRNA levels in association with obesity and diabetes were replicated in mice adipose tissue. In conclusion, this is the first observation, to our knowledge, of lactoferrin gene expression in whole adipose tissue and isolated adipocytes, increasing during adipogenesis and suggesting a possible contribution in adipose tissue physiology through LRP1.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Biomarkers/metabolism , Inflammation/metabolism , Lactoferrin/genetics , 3T3-L1 Cells , Adult , Animals , Female , Fibroblasts/metabolism , Humans , Male , Mice , Middle Aged
8.
J Mol Med (Berl) ; 91(4): 523-34, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23207880

ABSTRACT

Calgranulin B (S100A9) was recognized as a candidate type 2 diabetes (T2D) gene in the genomic profiling of muscle from a rodent model of T2D and identifying the human orthologs of genes localized in T2D susceptibility regions. Circulating and S100A9 expressions in muscle and adipose tissue, isolated fat cells, and mouse models were evaluated. A common 5'-upstream single-nucleotide polymorphism (SNP; rs3014866) for S100A9 was analyzed, as well as the effects of weight loss and treatments in vitro with recombinant S100A9. S100a9 expression was increased in muscle of diabetic mice (1.6-fold, p = 0.002), and in muscle from subjects with impaired glucose tolerance (∼4-fold, p = 0.028; n = 34). The rs3014866 SNP was associated with circulating S100A9 and the risk of T2D, having TT carriers at 28 % (p = 0.03) lower risk (n = 1,450). Indeed, increased circulating S100A9 (∼4-fold, p = 0.03; n = 206) and subcutaneous (2-fold, p = 0.01) and omental (1.4-fold, p = 0.04) S100A9 gene expressions (n = 83) in TT carriers run in parallel to decreased fasting glucose and glycated hemoglobin. Accordingly, metformin led to increased S100A9 mRNA in ex vivo-treated adipose tissue explants (n = 5/treatment). Otherwise, obese subjects showed a compensatory increase in circulating and S100A9 expressions in adipose (n = 126), as further demonstrated by decreased levels after diet- (-34 %, p = 0.002; n = 20) and surgery-induced (-58 %, p = 0.02; n = 8) weight loss. Lipopolysaccharide led to increased S100A9 in adipose from mice (n = 5/treatment) while recombinant S100A9 downregulated inflammation in adipocytes (n = 3/treatment). Current findings support the strategy of testing differentially expressed genes in mice and human orthologs associated with T2D. The increased S100A9 reported for obesity and insulin resistance may be envisioned as a compensatory mechanism for inflammation.


Subject(s)
Calgranulin B/genetics , Diabetes Mellitus, Type 2/genetics , Insulin Resistance/genetics , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adult , Aged , Alleles , Animals , Calgranulin B/blood , Calgranulin B/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Diet , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Genetic Association Studies , Genotype , Humans , Male , Metformin/pharmacology , Mice , Middle Aged , Muscles/drug effects , Muscles/metabolism , Polymorphism, Single Nucleotide
9.
Gut ; 61(4): 543-53, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22110050

ABSTRACT

OBJECTIVE: The gut microbiota, which is considered a causal factor in metabolic diseases as shown best in animals, is under the dual influence of the host genome and nutritional environment. This study investigated whether the gut microbiota per se, aside from changes in genetic background and diet, could sign different metabolic phenotypes in mice. METHODS: The unique animal model of metabolic adaptation was used, whereby C57Bl/6 male mice fed a high-fat carbohydrate-free diet (HFD) became either diabetic (HFD diabetic, HFD-D) or resisted diabetes (HFD diabetes-resistant, HFD-DR). Pyrosequencing of the gut microbiota was carried out to profile the gut microbial community of different metabolic phenotypes. Inflammation, gut permeability, features of white adipose tissue, liver and skeletal muscle were studied. Furthermore, to modify the gut microbiota directly, an additional group of mice was given a gluco-oligosaccharide (GOS)-supplemented HFD (HFD+GOS). RESULTS: Despite the mice having the same genetic background and nutritional status, a gut microbial profile specific to each metabolic phenotype was identified. The HFD-D gut microbial profile was associated with increased gut permeability linked to increased endotoxaemia and to a dramatic increase in cell number in the stroma vascular fraction from visceral white adipose tissue. Most of the physiological characteristics of the HFD-fed mice were modulated when gut microbiota was intentionally modified by GOS dietary fibres. CONCLUSIONS: The gut microbiota is a signature of the metabolic phenotypes independent of differences in host genetic background and diet.


Subject(s)
Adaptation, Physiological/physiology , Diet, High-Fat , Intestines/microbiology , Metagenome/physiology , Animals , Cecum/microbiology , Cytokines/blood , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/physiopathology , Fatty Acids, Nonesterified/blood , Glucose Tolerance Test , Intestinal Absorption/physiology , Lipopolysaccharides/blood , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Permeability , Phenotype
10.
Diabetes ; 60(8): 2179-86, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21700881

ABSTRACT

OBJECTIVE: The study objective was to evaluate the possible role of the macrophage molecule CD14 in insulin resistance. RESEARCH DESIGN AND METHODS: The effects of recombinant human soluble CD14 (rh-sCD14) on insulin sensitivity (clamp procedure) and adipose tissue gene expression were evaluated in wild-type (WT) mice, high fat-fed mice, ob/ob mice, and CD14 knockout (KO) mice. We also studied WT mice grafted with bone marrow stem cells from WT donor mice and CD14 KO mice. Finally, CD14 was evaluated in human adipose tissue and during differentiation of human preadipocytes. RESULTS: rh-sCD14 led to increased insulin action in WT mice, high-fat-fed mice, and ob/ob mice, but not in CD14 KO mice, in parallel to a marked change in the expression of 3,479 genes in adipose tissue. The changes in gene families related to lipid metabolism were most remarkable. WT mice grafted with bone marrow stem cells from WT donor mice became insulin resistant after a high-fat diet. Conversely, WT mice grafted with cells from CD14 KO mice resisted the occurrence of insulin resistance in parallel to decreased mesenteric adipose tissue inflammatory gene expression. Glucose intolerance did not worsen in CD14 KO mice grafted with bone marrow stem cells from high fat-fed WT mice when compared with recipient KO mice grafted with cells from CD14 KO donor mice. CD14 gene expression was increased in whole adipose tissue and adipocytes from obese humans and further increased after tumor necrosis factor-α. CONCLUSIONS: CD14 modulates adipose tissue inflammatory activity and insulin resistance.


Subject(s)
Inflammation/physiopathology , Insulin Resistance/physiology , Lipopolysaccharide Receptors/physiology , Adipocytes/cytology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Bone Marrow Transplantation , Cell Differentiation , Dietary Fats/administration & dosage , Gene Expression Profiling , Humans , Inflammation/immunology , Insulin Resistance/genetics , Lipopolysaccharide Receptors/genetics , Mice , Mice, Knockout , Mice, Obese , Tumor Necrosis Factor-alpha/pharmacology
11.
Front Biosci (Landmark Ed) ; 14(13): 5107-17, 2009 06 01.
Article in English | MEDLINE | ID: mdl-19482607

ABSTRACT

In humans, the intestinal microflora is inherited from our parents and from the environment. It has established an ecological mutualism with the host, allowing each organism to benefit from the symbiotic relationship. Based on recent evidence, some molecular mechanisms for the role of intestinal microflora on the control of energy metabolism have been proposed. During metabolic diseases such as obesity and diabetes, it has been proposed that an imbalance between the two dominant groups of beneficial bacteria, the Bacteroidetes and the Firmicutes, generates signals controlling the expression of genes by the epithelial intestinal cells. Genes involved in lipid metabolism such as the Fast Induced Adipocyte Factor have been considered as putative targets. In addition, bacterial extracts such as the lipopolysaccharides control the tone of the innate immune system thus regulating the general inflammatory status, insulin resistance, and adipose tissue plasticity. Therefore, strategies aimed at controlling the ecological mutualism between intestinal microflora and the host should lead to a new era of therapeutic and health benefits.


Subject(s)
Intestines/microbiology , Metabolic Diseases/therapy , Biological Evolution , Ecosystem , Energy Metabolism , Food/adverse effects , Humans , Inflammation/etiology , Insulin Resistance , Lipopolysaccharides/toxicity , Metabolic Diseases/etiology , Metagenome , Models, Biological , Probiotics/therapeutic use , Symbiosis , Weight Gain
SELECTION OF CITATIONS
SEARCH DETAIL
...