Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Obesity (Silver Spring) ; 28(6): 1086-1097, 2020 06.
Article in English | MEDLINE | ID: mdl-32281747

ABSTRACT

OBJECTIVE: Weight regain after weight loss is common, and there is evidence to suggest negative effects on health because of weight cycling. This study sought to investigate the impact of weight regain in formerly obese mice on adipose tissue architecture and stromal cell function. METHODS: A diet-switch model was employed for obesity induction, weight loss, and weight regain in mice. Flow cytometry quantified adipose tissue leukocytes in adipose tissue. Liver and adipose tissue depots were compared to determine tissue-specific effects of weight cycling. RESULTS: Epididymal white adipose tissue of formerly obese mice failed to expand in response to repeat exposure to high-fat diet and retained elevated numbers of macrophages and T cells. Weight regain was associated with disproportionally elevated liver mass, hepatic triglyceride content, serum insulin concentration, and serum transaminase concentration. These effects occurred despite an extended 6-month weight loss cycle and they demonstrate that formerly obese mice maintain durable alterations in their physiological response to weight regain. Conditioned media from epididymal adipose tissue of formerly obese mice inhibited adipogenesis of 3T3-L1 preadipocytes, suggesting a potential mechanism to explain failed epididymal adipose tissue expansion during weight regain. CONCLUSIONS: Metabolic abnormalities related to defects in adipose tissue expansion and ongoing dysfunction manifest in formerly obese mice during weight regain.


Subject(s)
Adipose Tissue/metabolism , Fatty Liver/metabolism , Obesity/metabolism , Weight Gain/physiology , Animals , Diet, High-Fat , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Obese
2.
Diabetes ; 66(2): 392-406, 2017 02.
Article in English | MEDLINE | ID: mdl-28108608

ABSTRACT

Obesity causes dramatic proinflammatory changes in the adipose tissue immune environment, but relatively little is known regarding how this inflammation responds to weight loss (WL). To understand the mechanisms by which meta-inflammation resolves during WL, we examined adipose tissue leukocytes in mice after withdrawal of a high-fat diet. After 8 weeks of WL, mice achieved similar weights and glucose tolerance values as age-matched lean controls but showed abnormal insulin tolerance. Despite fat mass normalization, total and CD11c+ adipose tissue macrophage (ATM) content remained elevated in WL mice for up to 6 months and was associated with persistent fibrosis in adipose tissue. ATMs in formerly obese mice demonstrated a proinflammatory profile, including elevated expression of interferon-γ, tumor necrosis factor-α, and interleukin-1ß. T-cell-deficient Rag1-/- mice showed a degree of ATM persistence similar to that in WT mice, but with reduced inflammatory gene expression. ATM proliferation was identified as the predominant mechanism by which ATMs are retained in adipose tissue with WL. Our study suggests that WL does not completely resolve obesity-induced ATM activation, which may contribute to the persistent adipose tissue damage and reduced insulin sensitivity observed in formerly obese mice.


Subject(s)
Adipose Tissue/immunology , Cell Proliferation , Macrophages/immunology , Obesity/immunology , Weight Loss/immunology , Adipose Tissue/cytology , Animals , Body Weight , Diet, High-Fat , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Glucose Tolerance Test , Homeodomain Proteins/genetics , Immunoblotting , Immunohistochemistry , Inflammation/immunology , Insulin/metabolism , Interferon-gamma/immunology , Interleukin-1beta/immunology , Macrophages/cytology , Male , Mice , Mice, Knockout , Mice, Obese , T-Lymphocytes , Tumor Necrosis Factor-alpha/immunology
3.
J Leukoc Biol ; 99(6): 1107-19, 2016 06.
Article in English | MEDLINE | ID: mdl-26658005

ABSTRACT

Obesity activates both innate and adaptive immune responses in adipose tissue, but the mechanisms critical for regulating these responses remain unknown. CD40/CD40L signaling provides bidirectional costimulatory signals between antigen-presenting cells and CD4(+) T cells, and CD40L expression is increased in obese humans. Therefore, we examined the contribution of CD40 to the progression of obesity-induced inflammation in mice. CD40 was highly expressed on adipose tissue macrophages in mice, and CD40/CD40L signaling promoted the expression of antigen-presenting cell markers in adipose tissue macrophages. When fed a high fat diet, Cd40-deficient mice had reduced accumulation of conventional CD4(+) T cells (Tconv: CD3(+)CD4(+)Foxp3(-)) in visceral fat compared with wild-type mice. By contrast, the number of regulatory CD4(+) T cells (Treg: CD3(+)CD4(+)Foxp3(+)) in lean and obese fat was similar between wild-type and knockout mice. Adipose tissue macrophage content and inflammatory gene expression in fat did not differ between obese wild-type and knockout mice; however, major histocompatibility complex class II and CD86 expression on adipose tissue macrophages was reduced in visceral fat from knockout mice. Similar results were observed in chimeric mice with hematopoietic Cd40-deficiency. Nonetheless, neither whole body nor hematopoietic disruption of CD40 ameliorated obesity-induced insulin resistance in mice. In human adipose tissue, CD40 expression was positively correlated with CD80 and CD86 expression in obese patients with type 2 diabetes. These findings indicate that CD40 signaling in adipose tissue macrophages regulates major histocompatibility complex class II and CD86 expression to control the expansion of CD4(+) T cells; however, this is largely dispensable for the development of obesity-induced inflammation and insulin resistance in mice.


Subject(s)
Adipose Tissue/pathology , CD4-Positive T-Lymphocytes/metabolism , CD40 Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Macrophages/metabolism , Obesity/immunology , Adiposity/drug effects , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD40 Ligand/metabolism , Diet, High-Fat , Hematopoiesis/drug effects , Humans , Insulin/pharmacology , Macrophages/drug effects , Male , Mice, Inbred C57BL , Obesity/pathology , Omentum/drug effects , Omentum/metabolism , Omentum/pathology , Signal Transduction/drug effects
4.
Adipocyte ; 4(4): 264-72, 2015.
Article in English | MEDLINE | ID: mdl-26451282

ABSTRACT

The extracellular matrix (ECM) plays important roles in maintaining adequate adipose tissue function and in metabolic regulation. Here we have examined the organization of a relatively unexplored adipose tissue ECM component, elastin and its response to diet induced obesity in mice. Additionally, we have explored the regulation and requirement of macrophage metalloelastase, MMP-12, in adipose tissue ECM remodeling in obesity. In visceral fat depots, elastin fibers form a mesh-like net that becomes denser with diet-induced obesity. In contrast, the elastin fibers in subcutaneous adipose depots are more linear in organization, and are tightly associated with adipose tissue macrophages (ATMs). We found that Mmp12 is produced predominantly by ATMs and can be induced with both short- and long-term high fat diet challenge and rapid remodeling induced by lipolysis. This contrasts with Mmp14 and Timp1 which are further induced only after chronic obesity in non-ATM populations. We examined obese transgenic Mmp12 (-/-) mice and found an increase in gene expression of ECM genes with diet-induced obesity, but showed few significant differences in metabolic parameters, elastin matrix density, or in adipose tissue inflammation. Together, these studies reveal the architecture and diet-induced regulation of the elastin matrix and suggest that MMP-12 is not required for elastin matrix remodeling or for the metabolic dysfunction that occurs with obesity.

5.
J Biol Chem ; 290(21): 13250-62, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25869128

ABSTRACT

Women of reproductive age are protected from metabolic disease relative to postmenopausal women and men. Most preclinical rodent studies are skewed toward the use of male mice to study obesity-induced metabolic dysfunction because of a similar protection observed in female mice. How sex differences in obesity-induced inflammatory responses contribute to these observations is unknown. We have compared and contrasted the effects of high fat diet-induced obesity on glucose metabolism and leukocyte activation in multiple depots in male and female C57Bl/6 mice. With both short term and long term high fat diet, male mice demonstrated increased weight gain and CD11c(+) adipose tissue macrophage content compared with female mice despite similar degrees of adipocyte hypertrophy. Competitive bone marrow transplant studies demonstrated that obesity induced a preferential contribution of male hematopoietic cells to circulating leukocytes and adipose tissue macrophages compared with female cells independent of the sex of the recipient. Sex differences in macrophage and hematopoietic cell in vitro activation in response to obesogenic cues were observed to explain these results. In summary, this report demonstrates that male and female leukocytes and hematopoietic stem cells have cell-autonomous differences in their response to obesity that contribute to an amplified response in males compared with females.


Subject(s)
Blood Glucose/metabolism , Diet, High-Fat/adverse effects , Hematopoietic Stem Cells/cytology , Inflammation/immunology , Obesity/etiology , Adipose Tissue/cytology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Animals , Biomarkers/analysis , Cells, Cultured , Colony-Forming Units Assay , Female , Flow Cytometry , Glucose Tolerance Test , Hematopoietic Stem Cells/metabolism , Immunohistochemistry , Inflammation/complications , Inflammation/pathology , Lipids/analysis , Male , Mice , Mice, Inbred C57BL , Myelopoiesis/physiology , Obesity/metabolism , Obesity/pathology , Sex Factors , Weight Gain
6.
Mol Metab ; 3(6): 664-75, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25161889

ABSTRACT

Obesity is associated with an activated macrophage phenotype in multiple tissues that contributes to tissue inflammation and metabolic disease. To evaluate the mechanisms by which obesity potentiates myeloid activation, we evaluated the hypothesis that obesity activates myeloid cell production from bone marrow progenitors to potentiate inflammatory responses in metabolic tissues. High fat diet-induced obesity generated both quantitative increases in myeloid progenitors as well as a potentiation of inflammation in macrophages derived from these progenitors. In vivo, hematopoietic stem cells from obese mice demonstrated the sustained capacity to preferentially generate inflammatory CD11c(+) adipose tissue macrophages after serial bone marrow transplantation. We identified that hematopoietic MyD88 was important for the accumulation of CD11c(+) adipose tissue macrophage accumulation by regulating the generation of myeloid progenitors from HSCs. These findings demonstrate that obesity and metabolic signals potentiate leukocyte production and that dietary priming of hematopoietic progenitors contributes to adipose tissue inflammation.

7.
PLoS One ; 8(3): e57929, 2013.
Article in English | MEDLINE | ID: mdl-23472120

ABSTRACT

Neuropeptide Y (NPY) is induced in peripheral tissues such as adipose tissue with obesity. The mechanism and function of NPY induction in fat are unclear. Given the evidence that NPY can modulate inflammation, we examined the hypothesis that NPY regulates the function of adipose tissue macrophages (ATMs) in response to dietary obesity in mice. NPY was induced by dietary obesity in the stromal vascular cells of visceral fat depots from mice. Surprisingly, the induction of Npy was limited to purified ATMs from obese mice. Significant basal production of NPY was observed in cultured bone marrow derived macrophage and dendritic cells (DCs) and was increased with LPS stimulation. In vitro, addition of NPY to myeloid cells had minimal effects on their activation profiles. NPY receptor inhibition promoted DC maturation and the production of IL-6 and TNFα suggesting an anti-inflammatory function for NPY signaling in DCs. Consistent with this, NPY injection into lean mice decreased the quantity of M1-like CD11c(+) ATMs and suppressed Ly6c(hi) monocytes. BM chimeras generated from Npy(-/-) donors demonstrated that hematopoietic NPY contributes to the obesity-induced induction of Npy in fat. In addition, loss of Npy expression from hematopoietic cells led to an increase in CD11c(+) ATMs in visceral fat with high fat diet feeding. Overall, our studies suggest that NPY is produced by a range of myeloid cells and that obesity activates the production of NPY in adipose tissue macrophages with autocrine and paracrine effects.


Subject(s)
Adipose Tissue/cytology , Gene Expression Regulation , Inflammation/metabolism , Macrophages/metabolism , Neuropeptide Y/metabolism , Obesity/metabolism , Animals , Dendritic Cells/cytology , Hematopoietic Stem Cells/cytology , Insulin Resistance , Interleukin-6/metabolism , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Monocytes/cytology , Receptors, Neuropeptide Y/metabolism , Triglycerides/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...