Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 557: 97-103, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33862466

ABSTRACT

Understanding of the mechanism of adipogenesis is essential for the control of obesity, which predisposes toward numerous health problems. High-mobility group box protein 2 (HMGB2) is a non-histone chromosomal protein that facilitates DNA replication, transcription, recombination, and repair. Here, we studied the role of HMGB2 in adipogenic differentiation. The expression of HMGB2 was measured at the mRNA and protein levels in cultured 3T3-L1 pre-adipocyte cells and during the process of adipogenic differentiation induced bya cocktail of insulin, 3-isobutyl-1-methylxanthine, and dexamethasone. This increased in the early phase and decreased in the late phase of differentiation. However, 3T3-L1 pre-adipocyte cells did not differentiate into adipocytes after the knockdown of HMGB2 expression by small interfering RNA (siRNA). Similarly, mesenchymal stem cells (MSCs) isolated from Hmgb2-/- mice did not express peroxisome proliferator-activated receptor gamma (PPARγ) in response to the adipocyte differentiation cocktail and did not differentiate. Wnt/ß-catenin signaling is a negative regulator of adipogenic differentiation. We found that ß-catenin expression was downregulated during 3T3-L1 adipogenic differentiation, as expected, but not when endogenous HMBG2 expression was knocked down using siRNA. These results indicate that HMGB2 plays an essential role in the early phase of the differentiation of pre-adipocytes and MSCs, and probably interacts with other regulators, such as PPARγ and Wnt/ß-catenin signaling.


Subject(s)
Adipocytes/cytology , Adipogenesis/physiology , HMGB2 Protein/metabolism , Mesenchymal Stem Cells/cytology , Wnt Signaling Pathway , Adipocytes/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Sci Rep ; 10(1): 4647, 2020 Mar 10.
Article in English | MEDLINE | ID: mdl-32157195

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Sci Rep ; 8(1): 9601, 2018 06 25.
Article in English | MEDLINE | ID: mdl-29942000

ABSTRACT

Although various surgical procedures have been developed for chronic rotator cuff tear repair, the re-tear rate remains high with severe fat infiltration. However, little is known about the molecular regulation of this process. Mesenchymal stem cells (MSCs) in the intra-muscular space are origin of ectopic fat cells in skeletal muscle. We have previously shown that high-mobility group box 2 (HMGB2), which is a nuclear protein commonly associated with mesenchymal differentiation, is involved in the early articular cartilage degeneration. In this study, we addressed the role of HMGB2 in adipogenesis of MSCs and fat infiltration into skeletal muscles. HMGB2 was highly expressed in undifferentiated MSCs and co-localized with platelet-derived growth factor receptor α (PDGFRA) known as an MSC-specific marker, while their expressions were decreased during adipocytic differentiation. Under the deficiency of HMGB2, the expressions of adipogenesis-related molecules were reduced, and adipogenic differentiation is substantially impaired in MSCs. Moreover, HMGB2+ cells were generated in the muscle belly of rat supraspinatus muscles after rotator cuff transection, and some of these cells expressed PDGFRA in intra-muscular spaces. Thus, our findings suggest that the enhance expression of HMGB2 induces the adipogenesis of MSCs and the fat infiltration into skeletal muscles through the cascade of HMGB2-PDGFRA.


Subject(s)
Adipogenesis , Adipose Tissue/cytology , Adipose Tissue/metabolism , HMGB2 Protein/metabolism , Muscle, Skeletal/cytology , Animals , Cell Differentiation , Gene Expression Regulation , HMGB2 Protein/genetics , Male , Mesenchymal Stem Cells/cytology , Mice , Rats , Receptors, Platelet-Derived Growth Factor/metabolism
4.
Drug Deliv ; 24(1): 558-568, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28181829

ABSTRACT

Our previous works demonstrated that brown rice-specific bioactive substance, γ-oryzanol acts as a chaperone, attenuates exaggerated endoplasmic reticulum (ER) stress in brain hypothalamus and pancreatic islets, thereby ameliorating metabolic derangement in high fat diet (HFD)-induced obese diabetic mice. However, extremely low absorption efficiency from intestine of γ-oryzanol is a tough obstacle for the clinical application. Therefore, in this study, to overcome extremely low bioavailability of γ-oryzanol with super-high lipophilicity, we encapsulated γ-oryzanol in polymer poly (DL-lactide-co-glycolide) (PLGA) nanoparticles (Nano-Orz), and evaluated its metabolically beneficial impact in genetically obese-diabetic ob/ob mice, the best-known severest diabetic model in mice. To our surprise, Nano-Orz markedly ameliorated fuel metabolism with an unexpected magnitude (∼1000-fold lower dose) compared with regular γ-oryzanol. Furthermore, such a conspicuous impact was achievable by its administration once every 2 weeks. Besides the excellent impact on dysfunction of hypothalamus and pancreatic islets, Nano-Orz markedly decreased ER stress and inflammation in liver and adipose tissue. Collectively, nanotechnology-based developments of functional foods oriented toward γ-oryzanol shed light on the novel approach for the treatment of a variety of metabolic diseases in humans.


Subject(s)
Diabetes Mellitus/drug therapy , Drug Carriers , Energy Metabolism/drug effects , Hypoglycemic Agents/administration & dosage , Hypolipidemic Agents/administration & dosage , Lactic Acid/administration & dosage , Nanoparticles , Obesity/drug therapy , Phenylpropionates/administration & dosage , Polyglycolic Acid/administration & dosage , Administration, Oral , Animals , Behavior, Animal/drug effects , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus/blood , Diabetes Mellitus/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Compounding , Endoplasmic Reticulum Stress/drug effects , Food Preferences/drug effects , Gastrointestinal Microbiome/drug effects , Hypoglycemic Agents/chemistry , Hypolipidemic Agents/chemistry , Insulin Resistance , Intestinal Absorption , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Lactic Acid/chemistry , Lipids/blood , Liver/drug effects , Liver/metabolism , Male , Mice, Obese , Nanomedicine , Obesity/blood , Obesity/genetics , Phenylpropionates/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Solubility , Technology, Pharmaceutical/methods , Time Factors
5.
Diabetes ; 64(4): 1120-30, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25315009

ABSTRACT

The current dogma is that obesity-associated hepatic inflammation is due to increased Kupffer cell (KC) activation. However, recruited hepatic macrophages (RHMs) were recently shown to represent a sizable liver macrophage population in the context of obesity. Therefore, we assessed whether KCs and RHMs, or both, represent the major liver inflammatory cell type in obesity. We used a combination of in vivo macrophage tracking methodologies and adoptive transfer techniques in which KCs and RHMs are differentially labeled with fluorescent markers. With these approaches, the inflammatory phenotype of these distinct macrophage populations was determined under lean and obese conditions. In vivo macrophage tracking revealed an approximately sixfold higher number of RHMs in obese mice than in lean mice, whereas the number of KCs was comparable. In addition, RHMs comprised smaller size and immature, monocyte-derived cells compared with KCs. Furthermore, RHMs from obese mice were more inflamed and expressed higher levels of tumor necrosis factor-α and interleukin-6 than RHMs from lean mice. A comparison of the MCP-1/C-C chemokine receptor type 2 (CCR2) chemokine system between the two cell types showed that the ligand (MCP-1) is more highly expressed in KCs than in RHMs, whereas CCR2 expression is approximately fivefold greater in RHMs. We conclude that KCs can participate in obesity-induced inflammation by causing the recruitment of RHMs, which are distinct from KCs and are not precursors to KCs. These RHMs then enhance the severity of obesity-induced inflammation and hepatic insulin resistance.


Subject(s)
Gluconeogenesis/physiology , Liver/metabolism , Macrophages/metabolism , Obesity/metabolism , Animals , Diet, High-Fat/adverse effects , Fatty Liver/metabolism , Fatty Liver/pathology , Interleukin-6/metabolism , Kupffer Cells/metabolism , Kupffer Cells/pathology , Liver/pathology , Macrophages/pathology , Male , Mice , Mice, Obese , Obesity/etiology , Obesity/pathology , Receptors, CCR2/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Diabetes ; 63(11): 3891-905, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24879833

ABSTRACT

Recently, pleiotropic benefits of incretin therapy beyond glycemic control have been reported. Although cancer is one of the main causes of death in diabetic patients, few reports describe the anticancer effects of incretin. Here, we examined the effect of the incretin drug exendin (Ex)-4, a GLP-1 receptor (GLP-1R) agonist, on prostate cancer. In human prostate cancer tissue obtained from patients after they had undergone radical prostatectomy, GLP-1R expression colocalized with P504S, a marker of prostate cancer. In in vitro experiments, Ex-4 significantly decreased the proliferation of the prostate cancer cell lines LNCap, PC3, and DU145, but not that of ALVA-41. This antiproliferative effect depended on GLP-1R expression. In accordance with the abundant expression of GLP-1R in LNCap cells, a GLP-1R antagonist or GLP-1R knockdown with small interfering RNA abolished the inhibitory effect of Ex-4 on cell proliferation. Although Ex-4 had no effect on either androgen receptor activation or apoptosis, it decreased extracellular signal-regulated kinase (ERK)-mitogen-activated protein kinase (MAPK) phosphorylation in LNCap cells. Importantly, Ex-4 attenuated in vivo prostate cancer growth induced by transplantation of LNCap cells into athymic mice and significantly reduced the tumor expression of P504S, Ki67, and phosphorylated ERK-MAPK. These data suggest that Ex-4 attenuates prostate cancer growth through the inhibition of ERK-MAPK activation.


Subject(s)
Peptides/pharmacology , Prostatic Neoplasms/metabolism , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Venoms/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Exenatide , Glucagon-Like Peptide-1 Receptor , Humans , Immunohistochemistry , In Vitro Techniques , Male , Mice , Mice, Nude , Receptors, Glucagon/antagonists & inhibitors
7.
Biochem Biophys Res Commun ; 450(1): 440-6, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24928396

ABSTRACT

In studies of gene-ablated mice, activin signaling through activin type IIB receptors (ActRIIB) and Smad2 has been shown to regulate not only pancreatic ß cell mass but also insulin secretion. However, it still remains unclear whether gain of function of activin signaling is involved in the modulation of pancreatic ß cell mass and insulin secretion. To identify distinct roles of activin signaling in pancreatic ß cells, the Cre-loxP system was used to activate signaling through activin type IB receptor (ActRIB) in pancreatic ß cells. The resultant mice (pancreatic ß cell-specific ActRIB transgenic (Tg) mice; ActRIBCAßTg) exhibited a defect in glucose-stimulated insulin secretion (GSIS) and a progressive impairment of glucose tolerance. Patch-clamp techniques revealed that the activity of ATP-sensitive K(+) channels (KATP channels) was decreased in mutant ß cells. These results indicate that an appropriate level of activin signaling may be required for GSIS in pancreatic ß cells, and that activin signaling involves modulation of KATP channel activity.


Subject(s)
Activin Receptors, Type I/metabolism , Activins/metabolism , Glucose/metabolism , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , KATP Channels/physiology , Animals , Cells, Cultured , Insulin Secretion , Ion Channel Gating/physiology , Mice , Mice, Transgenic , Signal Transduction/physiology
8.
Diabetologia ; 57(1): 157-66, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24068386

ABSTRACT

AIMS/HYPOTHESIS: The TGF-ß superfamily of ligands provides important signals for the development of pancreas islets. However, it is not yet known whether the TGF-ß family signalling pathway is required for essential islet functions in the adult pancreas. METHODS: To identify distinct roles for the downstream components of the canonical TGF-ß signalling pathway, a Cre-loxP system was used to disrupt SMAD2, an intracellular transducer of TGF-ß signals, in pancreatic beta cells (i.e. Smad2ß knockout [KO] mice). The activity of ATP-sensitive K(+) channels (KATP channels) was recorded in mutant beta cells using patch-clamp techniques. RESULTS: The Smad2ßKO mice exhibited defective insulin secretion in response to glucose and overt diabetes. Interestingly, disruption of SMAD2 in beta cells was associated with a striking islet hyperplasia and increased pancreatic insulin content, together with defective glucose-responsive insulin secretion. The activity of KATP channels was decreased in mutant beta cells. CONCLUSIONS/INTERPRETATION: These results suggest that in the adult pancreas, TGF-ß signalling through SMAD2 is crucial for not only the determination of beta cell mass but also the maintenance of defining features of mature pancreatic beta cells, and that this involves modulation of KATP channel activity.


Subject(s)
Hyperplasia/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , KATP Channels/metabolism , Smad2 Protein/metabolism , Animals , Electrophysiology , Female , Insulin Secretion , KATP Channels/genetics , Male , Mice , Mice, Knockout , Smad2 Protein/genetics
9.
Biochem Biophys Res Commun ; 436(3): 443-8, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23747729

ABSTRACT

Activin, a transforming growth factor ß family member, has a wide range of physiological roles during embryonic development and organogenesis. In the ovary, activin, secreted from ovarian granulosa cells, not only acts on the pituitary gland to regulate the gonadotropin secretion from the pituitary gland in an endocrine manner but also acts on granulosa cells in a paracrine/autocrine manner to regulate folliculogenesis. Previously, we showed that activin signals through activin type IB receptor (ActRIB) and up-regulates follicle-stimulating hormone receptor expression and P450 aromatase activity in human ovarian granulose cell-like KGN cells. In the current study, we demonstrate the direct involvement of Smad2 as a downstream signal mediator of ActRIB in the transcriptional regulation of the P450 aromatase gene (CYP19A) in KGN cells. Upon activin stimulation, Smad2 activation and an increase in P450 aromatase messenger RNA (mRNA) were observed in KGN cells. Interestingly, Smad2 phosphorylation correlated well with the increase in P450 aromatase mRNA. Reciprocally, knockdown of Smad2 mRNA in KGN cells led to a decrease in the P450 aromatase mRNA expression, suggesting that Smad2 regulates CYP19A gene expression. Further analysis of CYP19A promoter activity revealed that the 5' upstream region between -2069 and -1271bp is required for the activation by Smad2. Finally, we provide compelling evidence that Smad2 shows follicular stage-specific expression, which is high in granulosa cells of preantral or early antral follicles in mice. Our results suggest that activin signaling through the ActRIB-Smad2 pathway plays a pivotal role in CYP19A expression and thus in follicular development.


Subject(s)
Activin Receptors, Type I/metabolism , Activins/metabolism , Aromatase/metabolism , Gene Expression Regulation, Enzymologic , Granulosa Cells/enzymology , Smad2 Protein/metabolism , Activin Receptors, Type I/genetics , Animals , Aromatase/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Granulosa Cells/cytology , Humans , Mice , Phosphorylation , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Smad2 Protein/genetics , Transcriptional Activation
10.
Cell ; 153(2): 413-25, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23582329

ABSTRACT

Here, we demonstrate that the fractalkine (FKN)/CX3CR1 system represents a regulatory mechanism for pancreatic islet ß cell function and insulin secretion. CX3CR1 knockout (KO) mice exhibited a marked defect in glucose and GLP1-stimulated insulin secretion, and this defect was also observed in vitro in isolated islets from CX3CR1 KO mice. In vivo administration of FKN improved glucose tolerance with an increase in insulin secretion. In vitro treatment of islets with FKN increased intracellular Ca(2+) and potentiated insulin secretion in both mouse and human islets. The KO islets exhibited reduced expression of a set of genes necessary for the fully functional, differentiated ß cell state, whereas treatment of wild-type (WT) islets with FKN led to increased expression of these genes. Lastly, expression of FKN in islets was decreased by aging and high-fat diet/obesity, suggesting that decreased FKN/CX3CR1 signaling could be a mechanism underlying ß cell dysfunction in type 2 diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Receptors, Chemokine/metabolism , Signal Transduction , Adult , Aging , Animals , CX3C Chemokine Receptor 1 , Cadaver , Chemokine CX3CL1/administration & dosage , Chemokine CX3CL1/metabolism , Diet, High-Fat , Gene Expression , Glucose/metabolism , Humans , Hyperglycemia/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Receptors, Chemokine/genetics
11.
Endocrinology ; 154(6): 2188-99, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23525221

ABSTRACT

Female obesity is associated with insulin resistance, hyperandrogenemia, and reproductive dysfunction. We hypothesized that elevated free fatty acids (FFAs) might directly modulate pituitary gonadotropin production. FFAs caused a time- and dose-dependent increase in phosphorylation of the MAPKs p38MAPK, c-Jun N-terminal kinase (JNK)-1/2, and ERK1/2 in LßT2 gonadotrope cells. Furthermore, FFAs up-regulated Lhb mRNA expression acutely, an effect that was blocked by JNK inhibition, but suppressed Fshb mRNA expression, an effect that was independent of MAPK signaling. FFAs enhanced the activation of the MAPKs in the presence of GnRH, although the cotreatment did not alter Lhb induction but did eliminate the GnRH induction of Fshb. FFAs also suppressed activin-induced Fshb expression. Knockdown experiments showed that the FFA effect on the inflammatory kinases p38MAPK and JNK and on Lhb, but not Fshb, mRNA expression is mediated via toll-like receptor-2 and toll-like receptor-4 and was mimicked by lipopolysaccharide stimulation. In vivo, male C57BL/6 mice on a high-fat diet showed reduced FSH levels consistent with the suppression of Fshb seen in vitro. Histological analysis of the testes showed an increased number of abnormal seminiferous tubules. Female mice on a high-fat diet lacked the expected proestrus LH and FSH surge and exhibited an increase in the number of days at estrus and a reduced number of days at proestrus, and ovaries had significantly fewer corpora lutea. Taken together, our findings suggest that lipid excess can lead to reproductive defects in both male and female mice.


Subject(s)
Fatty Acids, Nonesterified/pharmacology , Gonadotrophs/drug effects , Obesity/metabolism , Proestrus/drug effects , RNA, Messenger/metabolism , Animals , Diet, High-Fat/adverse effects , Dose-Response Relationship, Drug , Female , Follicle Stimulating Hormone, beta Subunit/blood , Follicle Stimulating Hormone, beta Subunit/genetics , Follicle Stimulating Hormone, beta Subunit/metabolism , Gene Expression/drug effects , Gonadotrophs/cytology , Gonadotrophs/metabolism , Immunoblotting , Luteinizing Hormone, beta Subunit/blood , Luteinizing Hormone, beta Subunit/genetics , Luteinizing Hormone, beta Subunit/metabolism , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Obesity/etiology , Obesity/genetics , Ovary/drug effects , Ovary/metabolism , Pituitary Gland/cytology , Proestrus/genetics , Proestrus/metabolism , RNA Interference , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
12.
J Biol Chem ; 288(15): 10722-35, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23457303

ABSTRACT

Sirt1 is a NAD(+)-dependent class III deacetylase that functions as a cellular energy sensor. In addition to its well-characterized effects in peripheral tissues, emerging evidence suggests that neuronal Sirt1 activity plays a role in the central regulation of energy balance and glucose metabolism. To assess this idea, we generated Sirt1 neuron-specific knockout (SINKO) mice. On both standard chow and HFD, SINKO mice were more insulin sensitive than Sirt1(f/f) mice. Thus, SINKO mice had lower fasting insulin levels, improved glucose tolerance and insulin tolerance, and enhanced systemic insulin sensitivity during hyperinsulinemic euglycemic clamp studies. Hypothalamic insulin sensitivity of SINKO mice was also increased over controls, as assessed by hypothalamic activation of PI3K, phosphorylation of Akt and FoxO1 following systemic insulin injection. Intracerebroventricular injection of insulin led to a greater systemic effect to improve glucose tolerance and insulin sensitivity in SINKO mice compared with controls. In line with the in vivo results, insulin-induced AKT and FoxO1 phosphorylation were potentiated by inhibition of Sirt1 in a cultured hypothalamic cell line. Mechanistically, this effect was traced to a reduced effect of Sirt1 to directly deacetylate and repress IRS-1 function. The enhanced central insulin signaling in SINKO mice was accompanied by increased insulin receptor signal transduction in liver, muscle, and adipose tissue. In summary, we conclude that neuronal Sirt1 negatively regulates hypothalamic insulin signaling, leading to systemic insulin resistance. Interventions that reduce neuronal Sirt1 activity have the potential to improve systemic insulin action and limit weight gain on an obesigenic diet.


Subject(s)
Energy Metabolism/physiology , Hypothalamus/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Nerve Tissue Proteins/metabolism , Sirtuin 1/metabolism , Animals , Cells, Cultured , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Glucose/genetics , Glucose/metabolism , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Insulin/genetics , Insulin/pharmacology , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Organ Specificity , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sirtuin 1/genetics
13.
J Immunol ; 189(4): 1992-9, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22778393

ABSTRACT

GPR105, a G protein-coupled receptor for UDP-glucose, is highly expressed in several human tissues and participates in the innate immune response. Because inflammation has been implicated as a key initial trigger for type 2 diabetes, we hypothesized that GPR105 (official gene name: P2RY14) might play a role in the initiation of inflammation and insulin resistance in obesity. To this end, we investigated glucose metabolism in GPR105 knockout (KO) and wild-type (WT) mice fed a high-fat diet (HFD). We also examined whether GPR105 regulates macrophage recruitment to liver or adipose tissues by in vivo monocyte tracking and in vitro chemotaxis experiments, followed by transplantation of bone marrow from either KO or WT donors to WT recipients. Our data show that genetic deletion of GPR105 confers protection against HFD-induced insulin resistance, with reduced macrophage infiltration and inflammation in liver, and increased insulin-stimulated Akt phosphorylation in liver, muscle, and adipose tissue. By tracking monocytes from either KO or WT donors, we found that fewer KO monocytes were recruited to the liver of WT recipients. Furthermore, we observed that uridine 5-diphosphoglucose enhanced the in vitro migration of bone marrow-derived macrophages from WT but not KO mice, and that plasma uridine 5-diphosphoglucose levels were significantly higher in obese versus lean mice. Finally, we confirmed that insulin sensitivity improved in HFD mice with a myeloid cell-specific deletion of GPR105. These studies indicate that GPR105 ablation mitigates HFD-induced insulin resistance by inhibiting macrophage recruitment and tissue inflammation. Hence GPR105 provides a novel link between innate immunity and metabolism.


Subject(s)
Inflammation/metabolism , Insulin Resistance/immunology , Obesity/metabolism , Receptors, Purinergic P2/metabolism , Animals , Chemotaxis, Leukocyte/immunology , Diet, High-Fat/adverse effects , Flow Cytometry , Immunoblotting , Inflammation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/immunology , Receptors, Purinergic P2/immunology , Receptors, Purinergic P2Y , Reverse Transcriptase Polymerase Chain Reaction
14.
J Clin Invest ; 122(7): 2444-53, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22653059

ABSTRACT

Obesity-induced inflammation is a key component of systemic insulin resistance, which is a hallmark of type 2 diabetes. A major driver of this inflammation/insulin resistance syndrome is the accumulation of proinflammatory macrophages in adipose tissue and liver. We found that the orphan GPCR Gpr21 was highly expressed in the hypothalamus and macrophages of mice and that whole-body KO of this receptor led to a robust improvement in glucose tolerance and systemic insulin sensitivity and a modest lean phenotype. The improvement in insulin sensitivity in the high-fat diet-fed (HFD-fed) Gpr21 KO mouse was traced to a marked reduction in tissue inflammation caused by decreased chemotaxis of Gpr21 KO macrophages into adipose tissue and liver. Furthermore, mice lacking macrophage expression of Gpr21 were protected from HFD-induced inflammation and displayed improved insulin sensitivity. Results of in vitro chemotaxis studies in human monocytes suggested that the defect in chemotaxis observed ex vivo and in vivo in mice is also translatable to humans. Cumulatively, our data indicate that GPR21 has a critical function in coordinating macrophage proinflammatory activity in the context of obesity-induced insulin resistance.


Subject(s)
Diet, High-Fat/adverse effects , Insulin Resistance , Obesity/metabolism , Receptors, G-Protein-Coupled/genetics , Animals , Bone Marrow Transplantation , Eating , Energy Metabolism , Epididymis/metabolism , Gene Expression Profiling , Glucose/metabolism , Hypothalamus/metabolism , Inflammation Mediators/metabolism , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Liver/metabolism , Macrophages , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/pathology , Phenotype , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/metabolism , Sequence Deletion , Transcription, Genetic , Weight Gain
15.
J Biol Chem ; 287(28): 23852-63, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22532565

ABSTRACT

Lipid droplets (LDs) are ubiquitous organelles storing neutral lipids, including triacylglycerol (TAG) and cholesterol ester. The properties of LDs vary greatly among tissues, and LD-binding proteins, the perilipin family in particular, play critical roles in determining such diversity. Overaccumulation of TAG in LDs of non-adipose tissues may cause lipotoxicity, leading to diseases such as diabetes and cardiomyopathy. However, the physiological significance of non-adipose LDs in a normal state is poorly understood. To address this issue, we generated and characterized mice deficient in perilipin 5 (Plin5), a member of the perilipin family particularly abundant in the heart. The mutant mice lacked detectable LDs, containing significantly less TAG in the heart. Particulate structures containing another LD-binding protein, Plin2, but negative for lipid staining, remained in mutant mice hearts. LDs were recovered by perfusing the heart with an inhibitor of lipase. Cultured cardiomyocytes from Plin5-null mice more actively oxidized fatty acid than those of wild-type mice. Production of reactive oxygen species was increased in the mutant mice hearts, leading to a greater decline in heart function with age. This was, however, reduced by the administration of N-acetylcysteine, a precursor of an antioxidant, glutathione. Thus, we conclude that Plin5 is essential for maintaining LDs at detectable sizes in the heart, by antagonizing lipase(s). LDs in turn prevent excess reactive oxygen species production by sequestering fatty acid from oxidation and hence suppress oxidative burden to the heart.


Subject(s)
Fatty Acids/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Myocardium/metabolism , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Female , Free Radical Scavengers/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Lipase/metabolism , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Muscle Proteins/genetics , Myocardium/cytology , Myocardium/ultrastructure , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress , Triglycerides/metabolism
16.
Diabetes ; 61(2): 346-54, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22190646

ABSTRACT

Macrophage-mediated inflammation is a key component of insulin resistance; however, the initial events of monocyte migration to become tissue macrophages remain poorly understood. We report a new method to quantitate in vivo macrophage tracking (i.e., blood monocytes from donor mice) labeled ex vivo with fluorescent PKH26 dye and injected into recipient mice. Labeled monocytes appear as adipose, liver, and splenic macrophages, peaking in 1-2 days. When CCR2 KO monocytes are injected into wild-type (WT) recipients, or WT monocytes given to MCP-1 KO recipients, adipose tissue macrophage (ATM) accumulation is reduced by ~40%, whereas hepatic macrophage content is decreased by ~80%. Using WT donor cells, ATM accumulation is several-fold greater in obese recipient mice compared with lean mice, regardless of the source of donor monocytes. After their appearance in adipose tissue, ATMs progressively polarize from the M2- to the M1-like state in obesity. In summary, the CCR2/MCP-1 system is a contributory factor to monocyte migration into adipose tissue and is the dominant signal controlling the appearance of recruited macrophages in the liver. Monocytes from obese mice are not programmed to become inflammatory ATMs but rather the increased proinflammatory ATM accumulation in obesity is in response to tissue signals.


Subject(s)
Adipose Tissue/pathology , Macrophages/physiology , Obesity/pathology , Animals , CD11c Antigen/analysis , Cell Movement , Cell Polarity , Chemokine CCL2/physiology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Monocytes/physiology , Receptors, CCR2/physiology
17.
FASEB J ; 25(6): 1887-93, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21350118

ABSTRACT

Type 2 diabetes is highly prevalent in human populations, particularly in obese individuals, and is characterized by progressive pancreatic ß-cell dysfunction and insulin resistance. Most mammals, including Old World primates, express two major kinds of sialic acids, N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc), typically found at the distal ends of glycoconjugate chains at the cell surface. Humans are uniquely unable to produce endogenous Neu5Gc due to an inactivating mutation in the CMP-Neu5Ac hydroxylase (CMAH) gene. The CMAH enzyme catalyzes the generation of CMP-Neu5Gc by the transfer of a single oxygen atom to the acyl group of CMP-Neu5Ac. Here, we show that mice bearing a human-like deletion of the Cmah gene exhibit fasting hyperglycemia and glucose intolerance following a high-fat diet. This phenotype is caused not by worsened insulin resistance but by compromised pancreatic ß-cell function associated with a 65% decrease in islet size and area and 50% decrease in islet number. Obese Cmah-null mice also show an ∼40% reduction in response to insulin secretagogues in vivo. These findings show that human evolution-like changes in sialic acid composition impair pancreatic ß-cell function and exacerbate glucose intolerance in mice. This may lend insight into the pathogenesis of type 2 diabetes in obese humans.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/physiology , Mixed Function Oxygenases/genetics , Obesity/metabolism , Animals , Arginine/pharmacology , Blood Glucose , Diabetes Mellitus, Type 2/etiology , Dietary Fats/pharmacology , Glucose/pharmacology , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Hyperglycemia/genetics , Hyperglycemia/metabolism , Insulin-Secreting Cells/drug effects , Mice , Mice, Knockout , Mice, Obese , Mixed Function Oxygenases/deficiency , Mutation , Obesity/complications , Obesity/genetics
18.
EMBO J ; 29(24): 4223-36, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21045807

ABSTRACT

The macrophage-mediated inflammatory response is a key etiologic component of obesity-related tissue inflammation and insulin resistance. The transcriptional factor FoxO1 is a key regulator of cell metabolism, cell cycle and cell death. Its activity is tightly regulated by the phosphoinositide-3-kinase-AKT (PI3K-Akt) pathway, which leads to phosphorylation, cytoplasmic retention and inactivation of FoxO1. Here, we show that FoxO1 promotes inflammation by enhancing Tlr4-mediated signalling in mature macrophages. By means of chromatin immunoprecipitation (ChIP) combined with massively parallel sequencing (ChIP-Seq), we show that FoxO1 binds to multiple enhancer-like elements within the Tlr4 gene itself, as well as to sites in a number of Tlr4 signalling pathway genes. While FoxO1 potentiates Tlr4 signalling, activation of the latter induces AKT and subsequently inactivates FoxO1, establishing a self-limiting mechanism of inflammation. Given the central role of macrophage Tlr4 in transducing extrinsic proinflammatory signals, the novel functions for FoxO1 in macrophages as a transcriptional regulator of the Tlr4 gene and its inflammatory pathway, highlights FoxO1 as a key molecular adaptor integrating inflammatory responses in the context of obesity and insulin resistance.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Macrophages/immunology , Signal Transduction , Toll-Like Receptor 4/biosynthesis , Animals , Cell Line , Chromatin Immunoprecipitation , DNA/metabolism , Enhancer Elements, Genetic , Forkhead Box Protein O1 , Inflammation Mediators/metabolism , Mice , Protein Binding , Sequence Analysis, DNA
19.
Cell ; 142(5): 687-98, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20813258

ABSTRACT

Omega-3 fatty acids (omega-3 FAs), DHA and EPA, exert anti-inflammatory effects, but the mechanisms are poorly understood. Here, we show that the G protein-coupled receptor 120 (GPR120) functions as an omega-3 FA receptor/sensor. Stimulation of GPR120 with omega-3 FAs or a chemical agonist causes broad anti-inflammatory effects in monocytic RAW 264.7 cells and in primary intraperitoneal macrophages. All of these effects are abrogated by GPR120 knockdown. Since chronic macrophage-mediated tissue inflammation is a key mechanism for insulin resistance in obesity, we fed obese WT and GPR120 knockout mice a high-fat diet with or without omega-3 FA supplementation. The omega-3 FA treatment inhibited inflammation and enhanced systemic insulin sensitivity in WT mice, but was without effect in GPR120 knockout mice. In conclusion, GPR120 is a functional omega-3 FA receptor/sensor and mediates potent insulin sensitizing and antidiabetic effects in vivo by repressing macrophage-induced tissue inflammation.


Subject(s)
Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/metabolism , Insulin Resistance , Receptors, G-Protein-Coupled/metabolism , 3T3-L1 Cells , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/metabolism , Cell Line , Dietary Fats/metabolism , Dietary Supplements , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/metabolism , Macrophages/immunology , Mice , Mice, Knockout , Obesity/complications , Receptors, G-Protein-Coupled/genetics
20.
Nat Cell Biol ; 11(8): 958-66, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19578372

ABSTRACT

Mitochondrial morphology is dynamically controlled by a balance between fusion and fission. The physiological importance of mitochondrial fission in vertebrates is less clearly defined than that of mitochondrial fusion. Here we show that mice lacking the mitochondrial fission GTPase Drp1 have developmental abnormalities, particularly in the forebrain, and die after embryonic day 12.5. Neural cell-specific (NS) Drp1(-/-) mice die shortly after birth as a result of brain hypoplasia with apoptosis. Primary culture of NS-Drp1(-/-) mouse forebrain showed a decreased number of neurites and defective synapse formation, thought to be due to aggregated mitochondria that failed to distribute properly within the cell processes. These defects were reflected by abnormal forebrain development and highlight the importance of Drp1-dependent mitochondrial fission within highly polarized cells such as neurons. Moreover, Drp1(-/-) murine embryonic fibroblasts and embryonic stem cells revealed that Drp1 is required for a normal rate of cytochrome c release and caspase activation during apoptosis, although mitochondrial outer membrane permeabilization, as examined by the release of Smac/Diablo and Tim8a, may occur independently of Drp1 activity.


Subject(s)
Embryonic Development/physiology , GTP Phosphohydrolases/metabolism , Mitochondrial Proteins/metabolism , Synapses/enzymology , Animals , Animals, Newborn , Blotting, Western , Brain/cytology , Brain/embryology , Brain/metabolism , Cell Line , Cells, Cultured , Cytochromes c/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/ultrastructure , GTP Phosphohydrolases/genetics , Immunohistochemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Microscopy, Fluorescence , Mitochondria/enzymology , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Neurons/cytology , Neurons/metabolism , Synapses/metabolism , Synapses/ultrastructure , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...