Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters








Publication year range
1.
J Endocrinol ; 256(2)2023 02 01.
Article in English | MEDLINE | ID: mdl-36476560

ABSTRACT

Small molecule kinase inhibitors (SMKIs) are a class of therapeutic drugs that target protein kinases in diseases such as cancer. SMKIs are often designed to inhibit kinases involved in cell proliferation, but these drugs alter cell metabolism and the endocrine control of organismal metabolism. SMKI treatment in diabetic cancer patients reveals that certain SMKIs improve blood glucose levels and can mitigate insulin dependence or diabetic medication requirements in both type 1 diabetes (T1D) and type 2 diabetes (T2D). Certain SMKIs can preserve functional ß-cell mass and increase insulin secretion or insulin sensitivity. It is not yet clear why different SMKIs can have opposing effects on insulin and blood glucose. Understanding the therapeutic effects of these drugs in T1D and T2D is complicated by overlapping off-target effects of SMKIs. The potency of inhibition of the intended protein kinase and inhibition of multiple off-target kinases may underpin conflicting reports of how certain SMKIs alter blood glucose and insulin. We summarize the effects of SMKIs on the intended and off-target kinases that can alter blood glucose and insulin, including c-Abl, c-Kit, EGFR, and VEGF. Inhibition of PDGFRß consistently lowers blood glucose in T1D and T2D. The effects of SMKIs on the kinases that regulate immune pathways, such as BTK and RIPKs, mediate many of the diverse effects of these drugs on metabolism. We highlight that inhibition of RIPK2 by SMKIs is a central node in metabolism that influences key metabolic pathways including lipolysis, blood glucose control, insulin secretion, and insulin resistance.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Insulin Resistance , Neoplasms , Protein Kinase Inhibitors , Humans , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Insulin/metabolism , Insulin/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
2.
Physiol Rep ; 10(16): e15439, 2022 08.
Article in English | MEDLINE | ID: mdl-35993451

ABSTRACT

Postbiotics are microbial-derived components or metabolites that can influence host immunity and metabolism. Some postbiotics can improve blood glucose control and lower inflammation during bacterial or nutritional stress. Bacterial cell wall-derived muramyl dipeptide (MDP) is a potent insulin-sensitizing postbiotic that engages NOD2, RIPK2, and requires interferon regulatory factor 4 (IRF4) to lower inflammation and improve blood glucose. However, the sex-dependent effects of this postbiotic and the cell type required for IRF4 to cause inflammatory versus glycemic responses to MDP were unknown. Here, we measured how MDP injection altered glucose tolerance and adipose tissue inflammation during low-level endotoxemia and high fat diet (HFD)-induced obesity in male and female adipocyte-specific IRF4 knockout mice (AdipoIRF4fl/fl ) compared to WTfl/fl mice. Adipocyte IRF4 was required for the blood glucose-lowering effects of MDP during endotoxemia and HFD-induced obesity in male mice. However, MDP did not alter blood glucose in female WTfl/fl and AdipoIRF4fl/f mice during endotoxemia. Unexpectedly, female HFD-fed AdipoIRF4fl/f mice had lower blood glucose after MDP treatment compared to WTfl/fl mice. MDP lowered inflammatory gene expression in adipose tissue of HFD-fed WTfl/fl and AdipoIRF4fl/fl mice of both sexes. Therefore, MDP-mediated lowering of adipose inflammation does not require adipocyte IRF4 and was independent of sex. Together, these data show that injection of MDP, an insulin-sensitizing postbiotic, lowers adipose tissue inflammation in male and female mice, but lower adipose inflammation is not always associated with improved blood glucose. The blood glucose-lowering effect of the postbiotic MDP and dependence on adipocyte IRF4 is sex-dependent.


Subject(s)
Endotoxemia , Insulin Resistance , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Diet, High-Fat/adverse effects , Endotoxemia/complications , Female , Inflammation/metabolism , Insulin/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism
3.
Mol Metab ; 55: 101404, 2022 01.
Article in English | MEDLINE | ID: mdl-34839023

ABSTRACT

OBJECTIVE: Obesity and diabetes increase circulating levels of microbial components derived from the gut microbiota. Individual bacterial factors (i.e., postbiotics) can have opposing effects on blood glucose. METHODS: We tested the net effect of gut bacterial extracts on blood glucose in mice using a microbiota-based vaccination strategy. RESULTS: Male and female mice had improved glucose and insulin tolerance five weeks after a single subcutaneous injection of a specific dose of a bacterial extract obtained from the luminal contents of the upper small intestine (SI), lower SI, or cecum. Injection of mice with intestinal extracts from germ-free mice revealed that bacteria were required for a microbiota-based vaccination to improve blood glucose control. Vaccination of Nod1-/-, Nod2-/-, and Ripk2-/- mice showed that each of these innate immune proteins was required for bacterial extract injection to improve blood glucose control. A microbiota-based vaccination promoted an immunoglobulin-G (IgG) response directed against bacterial extract antigens, where subcutaneous injection of mice with the luminal contents of the lower SI elicited a bacterial extract-specific IgG response that is compartmentalized to the lower SI of vaccinated mice. A microbiota-based vaccination was associated with an altered microbiota composition in the lower SI and colon of mice. Lean mice only required a single injection of small intestinal-derived bacterial extract, but high fat diet (HFD)-fed, obese mice required prime-boost bacterial extract injections for improvements in blood glucose control. CONCLUSIONS: Subversion of the gut barrier by vaccination with a microbiota-based extract engages innate immunity to promote long-lasting improvements in blood glucose control in a dose-dependent manner.


Subject(s)
Blood Glucose/drug effects , Gastrointestinal Microbiome/immunology , Vaccination/methods , Animals , Cecum , Diabetes Mellitus , Diet, High-Fat , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Glucose/metabolism , Glycemic Control/methods , Immunity, Innate/immunology , Insulin Resistance/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Microbiota , Obesity/metabolism
4.
J Biol Chem ; 296: 100440, 2021.
Article in English | MEDLINE | ID: mdl-33610548

ABSTRACT

Obesity associates with inflammation, insulin resistance, and higher blood lipids. It is unclear if immune responses facilitate lipid breakdown and release from adipocytes via lipolysis in a separate way from hormones or adrenergic signals. We found that an ancient component of ER stress, inositol-requiring protein 1 (IRE1), discriminates inflammation-induced adipocyte lipolysis versus lipolysis from adrenergic or hormonal stimuli. Our data show that inhibiting IRE1 kinase activity was sufficient to block adipocyte-autonomous lipolysis from multiple inflammatory ligands, including bacterial components, certain cytokines, and thapsigargin-induced ER stress. IRE1-mediated lipolysis was specific for inflammatory triggers since IRE1 kinase activity was dispensable for isoproterenol and cAMP-induced lipolysis in adipocytes and mouse adipose tissue. IRE1 RNase activity was not associated with inflammation-induced adipocyte lipolysis. Inhibiting IRE1 kinase activity blocked NF-κB activation, interleukin-6 secretion, and adipocyte-autonomous lipolysis from inflammatory ligands. Inflammation-induced lipolysis mediated by IRE1 occurred independently from changes in insulin signaling in adipocytes, suggesting that inflammation can promote IRE1-mediated lipolysis independent of adipocyte insulin resistance. We found no role for canonical unfolded protein responses or ABL kinases in linking ER stress to IRE1-mediated lipolysis. Adiponectin-Cre-mediated IRE1 knockout in mice showed that adipocyte IRE1 was required for inflammatory ligand-induced lipolysis in adipose tissue explants and that adipocyte IRE1 was required for approximately half of the increase in blood triglycerides after a bacterial endotoxin-mediated inflammatory stimulus in vivo. Together, our results show that IRE1 propagates an inflammation-specific lipolytic program independent from hormonal or adrenergic regulation. Targeting IRE1 kinase activity may benefit metabolic syndrome and inflammatory lipid disorders.


Subject(s)
Adipocytes/metabolism , Lipolysis/physiology , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , 3T3-L1 Cells , Adipocytes/physiology , Adipose Tissue/metabolism , Animals , Cytokines/metabolism , Inflammation/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Macrophages/metabolism , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , NF-kappa B/metabolism , Obesity/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Signal Transduction
5.
Mol Metab ; 42: 101067, 2020 12.
Article in English | MEDLINE | ID: mdl-32860984

ABSTRACT

OBJECTIVE: Hyperinsulinemia can be both a cause and consequence of obesity and insulin resistance. Hyperinsulinemia can result from increased insulin secretion and/or reduced insulin clearance. While many studies have focused on mechanisms triggering insulin secretion during obesity, the triggers for changes in insulin clearance during obesity are less defined. In this study, we investigated the role of the microbiota in regulating insulin clearance during diet-induced obesity. METHODS: Blood glucose and insulin clearance were tested in conventional male mice treated with antibiotics and germ-free mice colonized with microbes from mice that were fed a control (chow) diet or an obesogenic high-fat diet (HFD). The composition of the fecal microbiota was analyzed using 16S rRNA sequencing. RESULTS: Short-term HFD feeding and aging did not alter insulin clearance in the mice. Oral antibiotics mitigated impaired blood insulin clearance in the mice fed an HFD for 12 weeks or longer. Germ-free mice colonized with microbes from HFD-fed donor mice had impaired insulin but not C-peptide clearance. Microbe-transmissible insulin clearance impairment was only observed in germ-free mice after more than 6 weeks post-colonization upon HFD feeding. Five bacterial taxa predicted >90% of the variance in insulin clearance. Mechanistically, impaired insulin clearance was associated with lower levels of hepatic Ceacam-1 but increased liver and skeletal muscle insulin-degrading enzyme (IDE) activity. CONCLUSIONS: Gut microbes regulate insulin clearance during diet-induced obesity. A small cluster of microbes or their metabolites may be targeted for mitigating defects in insulin clearance and hyperinsulinemia during the progression of obesity and type 2 diabetes.


Subject(s)
Gastrointestinal Microbiome/physiology , Insulin/metabolism , Obesity/microbiology , Animals , Blood Glucose/metabolism , Diet, High-Fat , Feces/microbiology , Glucose/metabolism , Hyperinsulinism/metabolism , Insulin Resistance/physiology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , RNA, Ribosomal, 16S
6.
Endocrinology ; 161(8)2020 08 01.
Article in English | MEDLINE | ID: mdl-32473019

ABSTRACT

Tyrosine kinase inhibitors (TKIs) used in cancer are also being investigated in diabetes. TKIs can improve blood glucose control in diabetic cancer patients, but the specific kinases that alter blood glucose or insulin are not clear. We sought to define the role of Receptor Interacting Serine/Threonine Kinase 2 (RIPK2) in mouse models of insulin resistance. We tested the TKI gefitinib, which inhibits RIPK2 activity, in wild-type (WT), Nod1-/-, Nod2-/-, and Ripk2-/- mice fed an obesogenic high-fat diet. Gefitinib lowered blood glucose during a glucose tolerance test (GTT) in a nucleotide-binding oligomerization domain (NOD)-RIPK2-independent manner in all obese mice. However, gefitinib lowered glucose-stimulated insulin secretion only in obese Ripk2-/- mice. Gefitinib had no effect on insulin secretion in obese WT, Nod1-/-, or Nod2-/- mice. Hence, genetic deletion of Ripk2 promoted the insulin-sensitizing potential of gefitinib, since this TKI lowered both blood glucose and insulin only in Ripk2-/- mice. Gefitinib did not alter the inflammatory profile of pancreas, adipose, liver, or muscle tissues in obese Ripk2-/- mice compared with obese WT mice. We also tested imatinib, a TKI that does not inhibit RIPK2 activity, in obese WT mice. Imatinib lowered blood glucose during a GTT, consistent with TKIs lowering blood glucose independently of RIPK2. However, imatinib increased glucose-stimulated insulin secretion during the glucose challenge. These data show that multiple TKIs lower blood glucose, where actions of TKIs on RIPK2 dictate divergent insulin responses, independent of tissue inflammation. Our data show that RIPK2 limits the insulin sensitizing effect of gefitinib, whereas imatinib increased insulin secretion.


Subject(s)
Insulin Secretion/drug effects , Insulin Secretion/genetics , Obesity/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2/physiology , Adiposity/drug effects , Adiposity/genetics , Animals , Blood Glucose/drug effects , Blood Glucose/genetics , Blood Glucose/metabolism , Diet, High-Fat , Gefitinib/pharmacology , Insulin/metabolism , Insulin Resistance/genetics , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Nod1 Signaling Adaptor Protein/physiology , Nod2 Signaling Adaptor Protein/physiology , Obesity/etiology , Obesity/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
7.
Am J Physiol Endocrinol Metab ; 318(4): E579-E585, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32101030

ABSTRACT

Defining the host receptors and metabolic consequences of bacterial components can help explain how the microbiome influences metabolic diseases. Bacterial peptidoglycans that activate nucleotide-binding oligomerization domain-containing (NOD)1 worsen glucose control, whereas NOD2 activation improves glycemia. Receptor-interacting serine/threonine-protein kinase 2 (RIPK2) is required for innate immunity instigated by NOD1 and NOD2. The role of RIPK2 in the divergent effects of NOD1 versus NOD2 on blood glucose was unknown. We found that whole body deletion of RIPK2 negated all effects of NOD1 or NOD2 activation on blood glucose during an acute, low level endotoxin challenge in mice. It was known that NOD1 in hematopoietic cells participates in insulin resistance and metabolic inflammation in obese mice. It was unknown if RIPK2 in hematopoietic cells is required for the glucose-lowering and anti-inflammatory effects of NOD2 activation. We hypothesized that RIPK2 in nonhematopoietic cells dictated the glycemic effects of NOD2 activation. We found that whole body deletion of RIPK2 prevented the glucose-lowering effects of repeated NOD2 activation that were evident during a glucose tolerance test (GTT) in high-fat diet (HFD)-fed wild-type (WT) mice. NOD2 activation lowered glucose during a GTT and lowered adipose tissue inflammation in mice with RIPK2 deleted in hematopoietic cells. We conclude that RIPK2 in nonhematopoietic cells mediates the glucose lowering and anti-inflammatory effects of NOD2-activating postbiotics. We propose a model where lipopolysaccharides and NOD1 ligands synergize in hematopoietic cells to promote insulin resistance but NOD2 activation in nonhematopoietic cells promotes RIPK2-dependent immune tolerance and lowering of inflammation and insulin resistance.


Subject(s)
Blood Glucose/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Microbiota , Nod2 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Activation, Metabolic , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Diet, High-Fat , Glucose Tolerance Test , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Nod1 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics
8.
Diabetes ; 68(7): 1441-1448, 2019 07.
Article in English | MEDLINE | ID: mdl-31010959

ABSTRACT

Statins lower cholesterol and adverse cardiovascular outcomes, but this drug class increases diabetes risk. Statins are generally anti-inflammatory. However, statins can promote inflammasome-mediated adipose tissue inflammation and insulin resistance through an unidentified immune effector. Statins lower mevalonate pathway intermediates beyond cholesterol, but it is unknown whether lower cholesterol underpins statin-mediated insulin resistance. We sought to define the mevalonate pathway metabolites and immune effectors that propagate statin-induced adipose insulin resistance. We found that LDL cholesterol lowering was dispensable, but statin-induced lowering of isoprenoids required for protein prenylation triggered NLRP3/caspase-1 inflammasome activation and interleukin-1ß (IL-1ß)-dependent insulin resistance in adipose tissue. Multiple statins impaired insulin action at the level of Akt/protein kinase B signaling in mouse adipose tissue. Providing geranylgeranyl isoprenoids or inhibiting caspase-1 prevented statin-induced defects in insulin signaling. Atorvastatin (Lipitor) impaired insulin signaling in adipose tissue from wild-type and IL-18-/- mice, but not IL-1ß-/- mice. Atorvastatin decreased cell-autonomous insulin-stimulated lipogenesis but did not alter lipolysis or glucose uptake in 3T3-L1 adipocytes. Our results show that statin lowering of prenylation isoprenoids activates caspase-1/IL-1ß inflammasome responses that impair endocrine control of adipocyte lipogenesis. This may allow the targeting of cholesterol-independent statin side effects on adipose lipid handling without compromising the blood lipid/cholesterol-lowering effects of statins.


Subject(s)
Adipose Tissue/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Insulin/pharmacology , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/drug effects , Animals , Atorvastatin/adverse effects , Caspase 1/metabolism , Inflammasomes/drug effects , Inflammasomes/metabolism , Insulin Resistance , Interleukin-1beta/metabolism , Lipogenesis/drug effects , Male , Mevalonic Acid/metabolism , Mice , Mice, Mutant Strains , Prenylation/drug effects
9.
Nat Commun ; 9(1): 4681, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30409977

ABSTRACT

The intestinal microbiota and insulin sensitivity are rapidly altered after ingestion of obesogenic diets. We find that changes in the composition of the fecal microbiota precede changes in glucose tolerance when mice are fed obesogenic, low fiber, high fat diets (HFDs). Antibiotics alter glycemia during the first week of certain HFDs, but antibiotics show a more robust improvement in glycemic control in mice with protracted obesity caused by long-term feeding of multiple HFDs. Microbiota transmissible dysglycemia and glucose intolerance only occur when germ-free mice are exposed to obesity-related microbes for more than 45 days. We find that sufficient host exposure time to microbiota derived from HFD-fed mice allows microbial factors to contribute to insulin resistance, independently from increased adiposity in mice. Our results are consistent with intestinal microbiota contributing to chronic insulin resistance and dysglycemia during prolonged obesity, despite rapid diet-induced changes in the taxonomic composition of the fecal microbiota.


Subject(s)
Insulin Resistance , Microbiota , Obesity/microbiology , Adiposity/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Diet, High-Fat , Dysbiosis/microbiology , Dysbiosis/pathology , Feces/microbiology , Feeding Behavior/drug effects , Glucose Intolerance/pathology , Mice, Inbred C57BL , Microbiota/drug effects , Time Factors
10.
Sci Rep ; 7(1): 1578, 2017 05 08.
Article in English | MEDLINE | ID: mdl-28484277

ABSTRACT

Inflammation underpins aspects of insulin resistance and dysglycemia. Microbiota-derived cell wall components such as muropeptides or endotoxin can trigger changes in host immunity and metabolism. Specific peptidoglycan motifs promote metabolic tissue inflammation, lipolysis and insulin resistance via Nucleotide-binding oligomerization domain-containing protein 1 (Nod1). Receptor-interacting serine/threonine-protein kinase 2 (Ripk2) mediates Nod1-induced immunity, but the role of Ripk2 in metabolism is ill-defined. We hypothesized that Ripk2 was required for Nod1-mediated inflammation, lipolysis and dysglycemia. This is relevant because certain tyrosine kinase inhibitors (TKIs) inhibit Ripk2 and there is clinical evidence of TKIs lowering inflammation and blood glucose. Here, we showed that only a subset of TKIs known to inhibit Ripk2 attenuated Nod1 ligand-mediated adipocyte lipolysis. TKIs that inhibit Ripk2 decreased cytokine responses induced by Nod1-activating peptidoglycan, but not endotoxin in both metabolic and immune cells. Pre-treatment of adipocytes or macrophages with the TKI gefitinib inhibited Nod1-induced Cxcl1 and Il-6 secretion. Furthermore, treatment of mice with gefitinib prevented Nod1-induced glucose intolerance in vivo. Ripk2 was required for these effects on inflammation and metabolism, since Nod1-mediated cytokine and blood glucose changes were absent in Ripk2-/- mice. Our data show that specific TKIs used in cancer also inhibit Nod1-Ripk2 immunometabolism responses indicative of metabolic disease.


Subject(s)
Blood Glucose/metabolism , Cell Wall/metabolism , Inflammation/pathology , Lipolysis/drug effects , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cytokines/metabolism , Endotoxins , Gefitinib/pharmacology , Insulin Resistance , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Models, Biological , NF-kappa B/metabolism , Peptides/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
11.
Cell Metab ; 25(5): 1063-1074.e3, 2017 May 02.
Article in English | MEDLINE | ID: mdl-28434881

ABSTRACT

Intestinal dysbiosis contributes to obesity and insulin resistance, but intervening with antibiotics, prebiotics, or probiotics can be limited by specificity or sustained changes in microbial composition. Postbiotics include bacterial components such as lipopolysaccharides, which have been shown to promote insulin resistance during metabolic endotoxemia. We found that bacterial cell wall-derived muramyl dipeptide (MDP) is an insulin-sensitizing postbiotic that requires NOD2. Injecting MDP lowered adipose inflammation and reduced glucose intolerance in obese mice without causing weight loss or altering the composition of the microbiome. MDP reduced hepatic insulin resistance during obesity and low-level endotoxemia. NOD1-activating muropeptides worsened glucose tolerance. IRF4 distinguished opposing glycemic responses to different types of peptidoglycan and was required for MDP/NOD2-induced insulin sensitization and lower metabolic tissue inflammation during obesity and endotoxemia. IRF4 was dispensable for exacerbated glucose intolerance via NOD1. Mifamurtide, an MDP-based drug with orphan drug status, was an insulin sensitizer at clinically relevant doses in obese mice.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/immunology , Insulin Resistance , Interferon Regulatory Factors/immunology , Obesity/complications , Obesity/microbiology , Animals , Endotoxemia/complications , Endotoxemia/immunology , Endotoxemia/microbiology , Inflammation/complications , Inflammation/immunology , Inflammation/microbiology , Mice, Inbred C57BL , Mice, Obese , Microbiota , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Obesity/immunology
12.
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
13.
Diabetes ; 63(11): 3742-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24917577

ABSTRACT

Statins reduce lipid levels and are widely prescribed. Statins have been associated with an increased incidence of type 2 diabetes, but the mechanisms are unclear. Activation of the NOD-like receptor family, pyrin domain containing 3 (NLRP3)/caspase-1 inflammasome, promotes insulin resistance, a precursor of type 2 diabetes. We showed that four different statins increased interleukin-1ß (IL-1ß) secretion from macrophages, which is characteristic of NLRP3 inflammasome activation. This effect was dose dependent, absent in NLRP3(-/-) mice, and prevented by caspase-1 inhibition or the diabetes drug glyburide. Long-term fluvastatin treatment of obese mice impaired insulin-stimulated glucose uptake in adipose tissue. Fluvastatin-induced activation of the NLRP3/caspase-1 pathway was required for the development of insulin resistance in adipose tissue explants, an effect also prevented by glyburide. Fluvastatin impaired insulin signaling in lipopolysaccharide-primed 3T3-L1 adipocytes, an effect associated with increased caspase-1 activity, but not IL-1ß secretion. Our results define an NLRP3/caspase-1-mediated mechanism of statin-induced insulin resistance in adipose tissue and adipocytes, which may be a contributing factor to statin-induced development of type 2 diabetes. These results warrant scrutiny of insulin sensitivity during statin use and suggest that combination therapies with glyburide, or other inhibitors of the NLRP3 inflammasome, may be effective in preventing the adverse effects of statins.


Subject(s)
Adipose Tissue/metabolism , Carrier Proteins/metabolism , Fatty Acids, Monounsaturated/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Indoles/therapeutic use , Inflammasomes/drug effects , Inflammasomes/metabolism , 3T3-L1 Cells , Adipose Tissue/drug effects , Animals , Carrier Proteins/genetics , Caspase 1/genetics , Caspase 1/metabolism , Fatty Acids, Monounsaturated/adverse effects , Fluvastatin , Glyburide/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Indoles/adverse effects , Insulin Resistance , Interleukin-1beta/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Obesity/drug therapy , Obesity/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL