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.
J Biol Chem ; 288(31): 22565-75, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23788643

ABSTRACT

Complement activation is implicated in the development of obesity and insulin resistance, and loss of signaling by the anaphylatoxin C3a prevents obesity-induced insulin resistance in mice. Here we have identified C1q in the classical pathway as required for activation of complement in response to high fat diets. After 8 weeks of high fat diet, wild-type mice became obese and developed glucose intolerance. This was associated with increased apoptotic cell death and accumulation of complement activation products (C3b/iC3b/C3c) in liver and adipose tissue. Previous studies have shown that high fat diet-induced apoptosis is dependent on Bid; here we report that Bid-mediated apoptosis was required for complement activation in adipose and liver. Although C1qa deficiency had no effect on high fat diet-induced apoptosis, accumulation of complement activation products and the metabolic complications of high fat diet-induced obesity were dependent on C1q. When wild-type mice were fed a high fat diet for only 3 days, hepatic insulin resistance was associated with the accumulation of C3b/iC3b/C3c in the liver. Mice deficient in C3a receptor were protected against this early high fat diet-induced hepatic insulin resistance, whereas mice deficient in the negative complement regulator CD55/DAF were more sensitive to the high fat diet. C1qa(-/-) mice were also protected from high fat diet-induced hepatic insulin resistance and complement activation. Evidence of complement activation was also detected in adipose tissue of obese women compared with lean women. Together, these studies reveal an important role for C1q in the classical pathway of complement activation in the development of high fat diet-induced insulin resistance.


Subject(s)
Dietary Fats/administration & dosage , Glucose/metabolism , Homeostasis , Insulin Resistance , Liver/metabolism , Membrane Glycoproteins/genetics , Receptors, Complement/genetics , Adipose Tissue/metabolism , Animals , Male , Mice , Mice, Inbred C57BL
2.
Alcohol Clin Exp Res ; 36(2): 214-22, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21895711

ABSTRACT

BACKGROUND: Adipose tissue is an important target for ethanol action. One important effect of ethanol is to reduce the secretion of adiponectin from adipocytes; this decrease is associated with lowered circulating adiponectin in rodent models of chronic ethanol feeding. Adiponectin is an insulin-sensitizing, anti-inflammatory adipokine; decreased adiponectin activity may contribute to tissue injury in response to chronic ethanol. Here, we investigated the role of cytochrome P450 2E1 (CYP2E1) and oxidative stress in the mechanism for impaired adiponectin secretion from adipocytes in response to ethanol. METHODS: Male Wistar rats were fed a liquid diet containing ethanol as 36% of calories or pair-fed a control diet for 4 weeks. 3T3-L1 adipocyte cultures, expressing CYP2E1 or not, were exposed to ethanol or 4-hydroxynonenal (4-HNE). RESULTS: Chronic ethanol feeding to rats suppressed the secretion of adiponectin from isolated epididymal adipocytes. Ethanol feeding induced the expression of CYP2E1 in adipocytes and increased markers of oxidative stress, including 4-HNE and protein carbonyls. Because adiponectin is posttranslationally processed in the endoplasmic reticulum and Golgi, we investigated the impact of ethanol on the redox status of high-density microsomes. Chronic ethanol decreased the ratio of reduced glutathione to oxidized glutathione (4.6:1, pair-fed; 2.9:1, ethanol-fed) in high-density microsomes isolated from rat epididymal adipose tissue. We next utilized the 3T3-L1 adipocyte-like cell model to interrogate the mechanisms for impaired adiponectin secretion. Culture of 3T3-L1 adipocytes overexpressing exogenous CYP2E1, but not those overexpressing antisense CYP2E1, with ethanol increased oxidative stress and impaired adiponectin secretion from intracellular pools. Consistent with a role of oxidative stress in impaired adiponectin secretion, challenge of 3T3-L1 adipocytes with 4-HNE also reduced adiponectin mRNA expression and secretion, without affecting intracellular adiponectin concentration. CONCLUSIONS: These data demonstrate that CYP2E1-dependent reactive oxygen species production in response to ethanol disrupts adiponectin secretion from adipocytes.


Subject(s)
Adipocytes/metabolism , Adiponectin/metabolism , Central Nervous System Depressants/pharmacology , Cytochrome P-450 CYP2E1/physiology , Ethanol/pharmacology , Oxidative Stress/drug effects , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Biotin/analogs & derivatives , Biotin/pharmacology , Blotting, Western , Cell Separation , Enzyme-Linked Immunosorbent Assay , Glutathione/metabolism , Immunohistochemistry , In Vitro Techniques , L-Lactate Dehydrogenase/blood , Male , Mice , Microsomes/drug effects , Microsomes/metabolism , Protein Carbonylation/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects
3.
J Biol Chem ; 286(41): 35989-35997, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21856753

ABSTRACT

Chronic, heavy alcohol exposure results in inflammation in adipose tissue, insulin resistance, and liver injury. Here we have identified a CYP2E1/Bid/C1q-dependent pathway that is activated in response to chronic ethanol and is required for the development of inflammation in adipose tissue. Ethanol feeding for 25 days to wild-type (C57BL/6J) mice increased expression of multiple markers of adipose tissue inflammation relative to pair-fed controls independent of increased body weight or adipocyte size. Ethanol feeding increased the expression of CYP2E1 in adipocytes, but not stromal vascular cells, in adipose tissue and Cyp2e1(-/-) mice were protected from adipose tissue inflammation in response to ethanol. Ethanol feeding also increased the number of TUNEL-positive nuclei in adipose tissue of wild-type mice but not in Cyp2e1(-/-) or Bid (-/-) mice. Apoptosis contributed to adipose inflammation, as the expression of multiple inflammatory markers was decreased in mice lacking the Bid-dependent apoptotic pathway. The complement protein C1q binds to apoptotic cells, facilitating their clearance and activating complement. Making use of C1q-deficient mice, we found that activation of complement via C1q provided the critical link between CYP2E1/Bid-dependent apoptosis and onset of adipose tissue inflammation in response to chronic ethanol. In summary, chronic ethanol increases CYP2E1 activity in adipose, leading to Bid-mediated apoptosis and activation of complement via C1q, finally resulting in adipose tissue inflammation. Taken together, these data identify a novel mechanism for the development of adipose tissue inflammation that likely contributes to the pathophysiological effects of ethanol.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Central Nervous System Depressants/adverse effects , Complement C1q/metabolism , Cytochrome P-450 CYP2E1/metabolism , Ethanol/adverse effects , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , BH3 Interacting Domain Death Agonist Protein/genetics , Central Nervous System Depressants/pharmacokinetics , Complement C1q/genetics , Cytochrome P-450 CYP2E1/genetics , Ethanol/pharmacology , Female , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...