Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Rep ; 36(11): 109691, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525353

ABSTRACT

Lipopolysaccharides (LPSs) can promote metabolic endotoxemia, which is considered inflammatory and metabolically detrimental based on Toll-like receptor (TLR)4 agonists, such as Escherichia coli-derived LPS. LPSs from certain bacteria antagonize TLR4 yet contribute to endotoxemia measured by endotoxin units (EUs). We found that E. coli LPS impairs gut barrier function and worsens glycemic control in mice, but equal doses of LPSs from other bacteria do not. Matching the LPS dose from R. sphaeroides and E. coli by EUs reveals that only E. coli LPS promotes dysglycemia and adipose inflammation, delays intestinal glucose absorption, and augments insulin and glucagon-like peptide (GLP)-1 secretion. Metabolically beneficial endotoxemia promoted by R. sphaeroides LPS counteracts dysglycemia caused by an equal dose of E. coli LPS and improves glucose control in obese mice. The concept of metabolic endotoxemia should be expanded beyond LPS load to include LPS characteristics, such as lipid A acylation, which dictates the effect of metabolic endotoxemia.


Subject(s)
Endotoxemia/etiology , Intestines/drug effects , Lipopolysaccharides/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Blood Glucose/analysis , Body Weight/drug effects , Endotoxemia/metabolism , Escherichia coli/metabolism , Glucagon-Like Peptide 1/blood , Glucose/metabolism , Insulin/blood , Intestines/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Obesity/pathology , Peptidoglycan/pharmacology , Rhodobacter sphaeroides/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/metabolism
2.
J Endocrinol ; 250(2): R1-R21, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34165440

ABSTRACT

Micronutrients influence hormone action and host metabolism. Dietary minerals, trace elements, and vitamins can alter blood glucose and cellular glucose metabolism, and several micronutrients are associated with the risk and progression of type 2 diabetes. Dietary components, microbes, and host immune, endocrine, and metabolic responses all interact in the intestine. There has been a focus on macronutrients modifying the host-microbe relationship in metabolic disease. Micronutrients are positioned to alter host-microbe symbiosis that participates in host endocrine control of glucose metabolism. Minerals and trace elements can alter the composition of the intestinal microbiota, gut barrier function, compartmentalized metabolic inflammation, cellular glucose transport, and endocrine control of glucose metabolism, including insulin and thyroid hormones. Dietary vitamins also influence the composition of the intestinal microbiota and vitamins can be biotransformed by gut microbes. Host-microbe regulation of vitamins can alter immunity, lipid and glucose metabolism, and cell fate and function of pancreatic beta cells. Causal effects of micronutrients in host-microbe metabolism are still emerging, and the mechanisms linking dietary excess or deficiency of specific micronutrients to changes in gut microbes directly linked to metabolic disease risk are not yet clear. Dietary fiber, fat, protein, and carbohydrates are key dietary factors that impact how microbes participate in host glucose metabolism. It is possible that micronutrient and microbiota-derived factors also participate in host-microbe responses that tip the balance in the endocrine control of host glucose metabolism. Dietary micronutrients should be considered, tested, and controlled in pre-clinical and clinical studies investigating host-microbe factors in metabolic diseases.


Subject(s)
Blood Glucose/drug effects , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Micronutrients/administration & dosage , Animals , Diabetes Mellitus, Type 2/microbiology , Diet , Endocrine System/physiology , Female , Glucose/metabolism , Glycemic Control , Humans , Insulin , Male , Metabolic Diseases/microbiology , Obesity/microbiology , Pregnancy , Vitamins/administration & dosage
3.
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
4.
Am J Physiol Endocrinol Metab ; 319(2): E305-E314, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32516028

ABSTRACT

Obesity promotes nonalcoholic fatty liver disease (NAFLD). The intestinal microbiota contributes to NAFLD progression through a gut-to-liver pathway that promotes inflammation and fibrosis. Gut microbiota-derived factors can travel to the liver and activate immune responses in liver resident cells to promote inflammation and NAFLD. Little is known about bacterial sensors or immune responses that can protect against NAFLD. We tested whether the bacterial cell wall sensor nucleotide-binding oligomerization domain-containing (NOD)2 protects against diet-induced NAFLD in mice. Whole body deletion of NOD2 exacerbated liver steatosis and fibrosis in mice fed a NAFLD-promoting diet. Mice with a hepatocyte-specific deletion of NOD2 (Nod2-/-HKO) also had higher liver steatosis and fibrosis compared with littermate wild-type mice (WT) fed a NAFLD-promoting diet. Hepatocyte-specific NOD2 deletion altered the composition of the gut microbiome. Nod2-/-HKO mice had increased relative abundance of Clostridiales and lower Erysipelotrichaceae among other changes in cecal bacteria compared with littermate WT mice. Hepatocyte-specific NOD2 deletion altered a transcriptional program of liver inflammation, metabolism, and fibrosis. Nod2-/-HKO mice had higher levels of transcripts involved in lipid and cholesterol metabolism. Nod2-/-HKO mice had higher transcript levels of transforming growth factor-ß and collagen isoforms, which coincided with higher levels of liver collagen compared with WT mice. These data show that bacterial cell wall sensing within hepatocytes can engage retrograde cross-talk from the liver to the gut, where liver immunity communicates with the gut to influence the intestinal host-microbe relationship during diet-induced NAFLD, and NOD2 within the hepatocyte confers protection from liver steatosis and fibrosis.


Subject(s)
Dysbiosis/physiopathology , Gastrointestinal Microbiome/physiology , Liver Cirrhosis/physiopathology , Liver/physiopathology , Nod2 Signaling Adaptor Protein/physiology , Non-alcoholic Fatty Liver Disease/physiopathology , Animals , Diet , Dysbiosis/prevention & control , Hepatocytes/chemistry , Hepatocytes/physiology , Liver Cirrhosis/etiology , Liver Cirrhosis/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod2 Signaling Adaptor Protein/deficiency , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/prevention & control , Receptor Cross-Talk
5.
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
6.
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
7.
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
8.
Physiol Rep ; 6(22): e13930, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30485705

ABSTRACT

Immune components can bridge inflammatory triggers to metabolic dysfunction. Scavenger receptors sense lipoproteins, but it is not clear how different scavenger receptors alter carbohydrate metabolism during obesity. Macrophage scavenger receptor 1 (MSR1) and macrophage receptor with collagenous structure (MARCO) are scavenger receptors that have been implicated in lipoprotein metabolism and cardiovascular disease. We assessed glucose control, tissue-specific insulin sensitivity, and inflammation in Msr1- and Marco-deficient mice fed with obesogenic diets. Compared to wild-type (WT) mice, Msr1-/- mice had worse blood glucose control that was only revealed after diet-induced obesity, not in lean mice. Obese Msr1-/- mice had worse insulin-stimulated glucose uptake in the adipose tissue, which occurred in the absence of overt differences in adipose inflammation compared to obese WT mice. Msr1 deletion worsened dysglycemia independently from bacterial cell wall insulin sensitizers, such as muramyl dipeptide. MARCO was dispensable for glycemic control in obese mice. Oral administration of the polysaccharide fucoidan worsened glucose control in obese WT mice, but fucoidan had no effect on glycemia in obese Msr1-/- mice. Therefore, MSR1 is a scavenger receptor responsible for changes in glucose control in response to the environmental ligand fucoidan. Given the interest in dietary supplements and natural products reducing inflammation or insulin resistance in metabolic disease during obesity, our results highlight the importance of understanding which ligand-receptor relationships promote versus those that protect against metabolic disease factors. Our results show that ligand or gene targeting of MSR1 exacerbates insulin resistance in obese mice.


Subject(s)
Insulin Resistance , Obesity/metabolism , Scavenger Receptors, Class A/metabolism , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Insulin/metabolism , Ligands , Male , Mice , Mice, Inbred C57BL , Polysaccharides/pharmacology , Scavenger Receptors, Class A/drug effects , Scavenger Receptors, Class A/genetics
9.
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
10.
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
11.
J Obes Metab Syndr ; 26(3): 161-171, 2017 Sep.
Article in English | MEDLINE | ID: mdl-31089513

ABSTRACT

Obesity is associated with increased risk of developing metabolic diseases such as type 2 diabetes. The origins of obesity are multi-factorial, but ultimately rooted in increased host energy accumulation or retention. The gut microbiota has been implicated in control of host energy balance and nutrient extraction from dietary sources. The microbiota also impacts host immune status and dysbiosis-related inflammation can augment insulin resistance, independently of obesity. Advances in microbial metagenomic analyses and directly manipulating bacterial-host models of obesity have contributed to our understanding of the relationship between gut bacteria and metabolic disease. Foodborne, or drug-mediated perturbations to the gut microbiota can increase metabolic inflammation, insulin resistance, and dysglycemia. There is now some evidence that specific bacterial species can influence obesity and related metabolic defects such as insulin sensitivity. Components of bacteria are sufficient to impact obesity-related changes in metabolism. In fact, different microbial components derived from the bacterial cell wall can increase or decrease insulin resistance. Improving our understanding of the how components of the microbiota alter host metabolism is positioned to aid in the development of dietary interventions, avoiding triggers of dysbiosis, and generating novel therapeutic strategies to combat increasing rates of obesity and diabetes.

12.
Gut Microbes ; 7(1): 82-9, 2016.
Article in English | MEDLINE | ID: mdl-26939856

ABSTRACT

Microbes modify immunometabolism responses linking obesity and type 2 diabetes. Immunity helps maintain a host-microbe symbiosis, but inflammation can promote insulin resistance in tissues that control blood glucose. We were interested in compartmentalization of immune responses during obesity and show here that feeding mice an obesity-causing high-fat diet (HFD) decreased a marker of neutrophil activation and cytokines related to Th17 responses in the gut. A HFD decreased IL-17 and IL-21/22 in the ileum and colon, respectively. A HFD increased IL-17, IL-21/22 and other related Th17 responses in the liver. At the whole tissue level, there is divergence in gut and metabolic tissue Th17 cytokines during diet-induced obesity. Deletion of the bacterial peptidoglycan sensor NOD2 had relatively minor effects on these immune responses. We propose a model where diet-induced obesity promotes a permissive gut immune environment and sets the stage for host genetics to contribute to dysbiosis-driven metabolic tissue inflammation.


Subject(s)
Adipose Tissue/immunology , Diet, High-Fat , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Liver/immunology , Nod2 Signaling Adaptor Protein/deficiency , Obesity/immunology , Th17 Cells/immunology , Animals , Cytokines/immunology , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/microbiology , Male , Mice , Mice, Inbred C57BL , Neutrophil Activation/immunology , Neutrophils/immunology , Nod2 Signaling Adaptor Protein/genetics , Obesity/microbiology
13.
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
14.
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
15.
PLoS One ; 9(5): e97675, 2014.
Article in English | MEDLINE | ID: mdl-24828250

ABSTRACT

Obesity is associated with inflammation that can drive metabolic defects such as hyperlipidemia and insulin resistance. Specific metabolites can contribute to inflammation, but nutrient intake and obesity are also associated with altered bacterial load in metabolic tissues (i.e. metabolic endotoxemia). These bacterial cues can contribute to obesity-induced inflammation. The specific bacterial components and host receptors that underpin altered metabolic responses are emerging. We previously showed that Nucleotide-binding oligomerization domain-containing protein 1 (NOD1) activation with bacterial peptidoglycan (PGN) caused insulin resistance in mice. We now show that PGN induces cell-autonomous lipolysis in adipocytes via NOD1. Specific bacterial PGN motifs stimulated lipolysis in white adipose tissue (WAT) explants from WT, but not NOD1⁻/⁻mice. NOD1-activating PGN stimulated mitogen activated protein kinases (MAPK),protein kinase A (PKA), and NF-κB in 3T3-L1 adipocytes. The NOD1-mediated lipolysis response was partially reduced by inhibition of ERK1/2 or PKA alone, but not c-Jun N-terminal kinase (JNK). NOD1-stimulated lipolysis was partially dependent on NF-κB and was completely suppressed by inhibiting ERK1/2 and PKA simultaneously or hormone sensitive lipase (HSL). Our results demonstrate that bacterial PGN stimulates lipolysis in adipocytes by engaging a stress kinase, PKA, NF-κB-dependent lipolytic program. Bacterial NOD1 activation is positioned as a component of metabolic endotoxemia that can contribute to hyperlipidemia, systemic inflammation and insulin resistance by acting directly on adipocytes.


Subject(s)
Adipocytes/drug effects , Adipose Tissue/drug effects , Lipolysis/drug effects , Nod1 Signaling Adaptor Protein/genetics , Peptidoglycan/pharmacology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Gonads/cytology , Gonads/drug effects , Gonads/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/deficiency , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Tissue Culture Techniques
16.
Antimicrob Agents Chemother ; 57(7): 3078-84, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23612194

ABSTRACT

Pseudomonas aeruginosa is a leading cause of hospital-acquired infections and is resistant to many antibiotics. Among its primary mechanisms of resistance is expression of a chromosomally encoded AmpC ß-lactamase that inactivates ß-lactams. The mechanisms leading to AmpC expression in P. aeruginosa remain incompletely understood but are intricately linked to cell wall metabolism. To better understand the roles of peptidoglycan-active enzymes in AmpC expression-and consequent ß-lactam resistance-a phenotypic screen of P. aeruginosa mutants lacking such enzymes was performed. Mutants lacking one of four lytic transglycosylases (LTs) or the nonessential penicillin-binding protein PBP4 (dacB) had altered ß-lactam resistance. mltF and slt mutants with reduced ß-lactam resistance were designated WIMPs (wall-impaired mutant phenotypes), while highly resistant dacB, sltB1, and mltB mutants were designated HARMs (high-level AmpC resistant mutants). Double mutants lacking dacB and sltB1 had extreme piperacillin resistance (>256 µg/ml) compared to either of the single knockouts (64 µg/ml for a dacB mutant and 12 µg/ml for an sltB1 mutant). Inactivation of ampC reverted these mutants to wild-type susceptibility, confirming that AmpC expression underlies resistance. dacB mutants had constitutively elevated AmpC expression, but the LT mutants had wild-type levels of AmpC in the absence of antibiotic exposure. These data suggest that there are at least two different pathways leading to AmpC expression in P. aeruginosa and that their simultaneous activation leads to extreme ß-lactam resistance.


Subject(s)
Bacterial Proteins/metabolism , Peptidoglycan/metabolism , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/enzymology , beta-Lactam Resistance/genetics , beta-Lactamases/metabolism , Glycosyltransferases/genetics , Microbial Sensitivity Tests , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/metabolism , Pseudomonas aeruginosa/genetics , beta-Lactams/pharmacology
17.
PLoS One ; 8(3): e60666, 2013.
Article in English | MEDLINE | ID: mdl-23544160

ABSTRACT

Active efflux of antimicrobial agents is a primary mechanism by which bacterial pathogens can become multidrug resistant. The combined use of efflux pump inhibitors (EPIs) with pump substrates is under exploration to overcome efflux-mediated multidrug resistance. Phenylalanine-arginine ß-naphthylamide (PAßN) is a well-studied EPI that is routinely combined with fluoroquinolone antibiotics, but few studies have assessed its utility in combination with ß-lactam antibiotics. The initial goal of this study was to assess the efficacy of ß-lactams in combination with PAßN against the opportunistic pathogen, Pseudomonas aeruginosa. PAßN reduced the minimal inhibitory concentrations (MICs) of several ß-lactam antibiotics against P. aeruginosa; however, the susceptibility changes were not due entirely to efflux inhibition. Upon PAßN treatment, intracellular levels of the chromosomally-encoded AmpC ß-lactamase that inactivates ß-lactam antibiotics were significantly reduced and AmpC levels in supernatants correspondingly increased, potentially due to permeabilization of the outer membrane. PAßN treatment caused a significant increase in uptake of 8-anilino-1-naphthylenesulfonic acid, a fluorescent hydrophobic probe, and sensitized P. aeruginosa to bulky antibiotics (e.g. vancomycin) that are normally incapable of crossing the outer membrane, as well as to detergent-like bile salts. Supplementation of growth media with magnesium to stabilize the outer membrane increased MICs in the presence of PAßN and restored resistance to vancomycin. Thus, PAßN permeabilizes bacterial membranes in a concentration-dependent manner at levels below those typically used in combination studies, and this additional mode of action should be considered when using PAßN as a control for efflux studies.


Subject(s)
Cell Membrane Permeability/drug effects , Dipeptides/pharmacology , Gram-Negative Bacteria/cytology , Gram-Negative Bacteria/drug effects , Anilino Naphthalenesulfonates/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Culture Media , Detergents/pharmacology , Drug Resistance, Bacterial/drug effects , Magnesium/pharmacology , Microbial Sensitivity Tests , Periplasm/drug effects , Periplasm/metabolism , Pseudomonas aeruginosa/drug effects , beta-Lactamases/metabolism , beta-Lactams/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...