Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Lipids Health Dis ; 11: 21, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22313574

ABSTRACT

BACKGROUND: Mast cells are implicated in the pathogenesis of obesity and insulin resistance. Here, we explored the effects of leptin deficiency-induced obesity on the density of mast cells in metabolic (abdominal fat depots, skeletal muscle, and liver) and lymphatic (abdominal lymph nodes, spleen, and thymus) organs. Fourteen-week-old male leptin-deficient ob/ob mice and their controls fed a standard chow were studied. Tissue sections were stained with toluidine blue to determine the density of mast cells. CD117/c-kit protein expression analysis was also carried out. Furthermore, mast cells containing immunoreactive tumor necrosis factor-α (TNF-α), a proinflammatory cytokine involved in obesity-linked insulin resistance, were identified by immunostaining. RESULTS: ob/ob mice demonstrated adiposity and insulin resistance. In abdominal fat depots, mast cells were distributed differentially. While most prevalent in subcutaneous fat in controls, mast cells were most abundant in epididymal fat in ob/ob mice. Leptin deficiency-induced obesity was accompanied by a 20-fold increase in the density of mast cells in epididymal fat, but a 13-fold decrease in subcutaneous fat. This finding was confirmed by CD117/c-kit protein expression analysis. Furthermore, we found that a subset of mast cells in epididymal and subcutaneous fat were immunoreactive for TNF-α. The proportion of mast cells immunoreactive for TNF-α was higher in epididymal than in subcutaneous fat in both ob/ob and control mice. Mast cells were also distributed differentially in retroperitoneal, mesenteric, and inguinal lymph nodes. In both ob/ob mice and lean controls, mast cells were more prevalent in retroperitoneal than in mesenteric and inguinal lymph nodes. Leptin deficiency-induced obesity was accompanied by increased mast cell density in all lymph node stations examined. No significant difference in the density of mast cells in skeletal muscle, liver, spleen, and thymus was noted between ob/ob and control mice. CONCLUSIONS: This study demonstrates that leptin deficiency-induced obesity is accompanied by alterations in the density of mast cells in abdominal fat depots. The divergent distribution of mast cells in subcutaneous versus visceral fat might partially account for their differential biological behavior. Mast cells might also play a role in adaptive immune response occurring in regional lymph nodes in obesity.


Subject(s)
Abdominal Fat/pathology , Leptin/deficiency , Lymph Nodes/pathology , Mast Cells/physiology , Obesity/pathology , Adiposity , Animals , Blood Glucose , Cell Count , Cholesterol/blood , Epididymis/immunology , Epididymis/pathology , Liver/pathology , Male , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Obese , Muscle, Skeletal/pathology , Obesity/blood , Obesity/etiology , Organ Specificity , Proto-Oncogene Proteins c-kit , Spleen/pathology , Subcutaneous Fat/immunology , Subcutaneous Fat/pathology , Thymus Gland/pathology , Tumor Necrosis Factor-alpha/immunology
2.
Lipids Health Dis ; 10: 198, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-22051061

ABSTRACT

BACKGROUND: Obesity is characterized by increased cell death and inflammatory reactions in the adipose tissue. Here, we explored pathophysiological alterations taking place in the adipose tissue in long-standing obesity. In the epididymal fat of C57BL/6 mice fed a high-fat diet for 20 weeks, the prevalence and distribution of dead adipocytes (crown-like structures), mast cells (toluidine blue, mMCP6), macrophages (F4/80), and apoptotic cells (cleaved caspase-3) were measured. Moreover, gene and/or protein expression of several adipocytokines (leptin, adiponectin, TNF-α, IL-10, IL-6, MCP-1), F4/80, mMCP6, cleaved caspase-3 were determined. RESULTS: We observed that the epididymal fat mass was lower in obese than in lean mice. In obese mice, the epididymal fat mass correlated inversely with body weight and liver mass. Dead adipocytes, mast cells, macrophages, and apoptotic cells were abundant in the epididymal fat of obese mice, especially in the rostral vs. caudal zone. Accordingly, mMCP6, F4/80, and cleaved caspase-3 gene and/or protein expression was increased. Conversely, adiponectin, leptin, IL-6, and MCP-1 gene expression levels were lower in the epididymal fat of obese than lean mice. Although TNF-α and IL-10 gene expression was higher in the epididymal fat of obese mice, their expression relative to F4/80 and mMCP6 expression were lower in the heavily infiltrated rostral than caudal zone. CONCLUSIONS: This study demonstrates that in mice with long-standing obesity diminished gene expression of several adipocytokines accompany apoptosis and reduced mass of the epididymal fat. Our findings suggest that this is due to both increased prevalence of dead adipocytes and altered immune cell activity. Differential distribution of metabolically challenged adipocytes is indicative of the presence of biologically diverse zones within the epididymal fat.


Subject(s)
Adiponectin/genetics , Adiposity , Leptin/genetics , Obesity/pathology , Adiponectin/metabolism , Adipose Tissue/pathology , Animals , Apoptosis , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Diet, High-Fat , Down-Regulation , Epididymis/enzymology , Epididymis/metabolism , Gene Expression , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Leptin/metabolism , Liver/pathology , Macrophages/pathology , Male , Mast Cells/pathology , Mastocytosis , Mice , Mice, Inbred C57BL , Obesity/metabolism , Organ Size , Organ Specificity , Random Allocation , Tryptases/genetics , Tryptases/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
3.
J Lipid Res ; 52(3): 480-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21148461

ABSTRACT

Obesity is accompanied by adipocyte death and accumulation of macrophages and mast cells in expanding adipose tissues. Considering the differences in biological behavior of fat found in different anatomical locations, we explored the distribution of mast cells, solitary macrophages, and crown-like structures (CLS), the surrogates for dead adipocytes, in subcutaneous and abdominal visceral fat of lean and diet-induced obese C57BL/6 mice. In fat depots of lean mice, mast cells were far less prevalent than solitary macrophages. Subcutaneous fat contained more mast cells, but fewer solitary macrophages and CLS, than visceral fat. Whereas no significant change in mast cell density of subcutaneous fat was observed, obesity was accompanied by a substantial increase in mast cells in visceral fat. CLS became prevalent in visceral fat of obese mice, and the distribution paralleled mast cells. Adipose tissue mast cells contained and released preformed TNF-α, the cytokine implicated in the pathogenesis of obesity-linked insulin resistance. In summary, subcutaneous fat differed from visceral fat by immune cell composition and a lower prevalence of CLS both in lean and obese mice. The increase in mast cells in visceral fat of obese mice suggests their role in the pathogenesis of obesity and insulin resistance.


Subject(s)
Intra-Abdominal Fat/pathology , Macrophages/pathology , Mast Cells/pathology , Subcutaneous Fat/pathology , Animals , Diet/adverse effects , Epididymis/pathology , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/pathology , Thinness/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...