Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(17): e2401716121, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38625937

ABSTRACT

Serine phosphorylations on insulin receptor substrate 1 (IRS-1) by diverse kinases aoccur widely during obesity-, stress-, and inflammation-induced conditions in models of insulin resistance and type 2 diabetes. In this study, we define a region within the human IRS-1, which is directly C-terminal to the PTB domain encompassing numerous serine phosphorylation sites including Ser307 (mouse Ser302) and Ser312 (mouse 307) creating a phosphorylation insulin resistance (PIR) domain. We demonstrate that the IRS-1 PTB-PIR with its unphosphorylated serine residues interacts with the insulin receptor (IR) but loses the IR-binding when they are phosphorylated. Surface plasmon resonance studies further confirm that the PTB-PIR binds stronger to IR than just the PTB domain, and that phosphorylations at Ser307, Ser312, Ser315, and Ser323 within the PIR domain result in abrogating the binding. Insulin-responsive cells containing the mutant IRS-1 with all these four serines changed into glutamates to mimic phosphorylations show decreased levels of phosphorylations in IR, IRS-1, and AKT compared to the wild-type IRS-1. Hydrogen-deuterium exchange mass spectrometry experiments indicating the PIR domain interacting with the N-terminal lobe and the hinge regions of the IR kinase domain further suggest the possibility that the IRS-1 PIR domain protects the IR from the PTP1B-mediated dephosphorylation.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Mice , Humans , Animals , Phosphorylation , Serine/metabolism , Receptor, Insulin/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Cell Line , Phosphoproteins/metabolism , Insulin/metabolism
2.
Mol Cells ; 47(2): 100031, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38354858

ABSTRACT

It is now well-accepted that obesity-induced inflammation plays an important role in the development of insulin resistance and type 2 diabetes. A key source of the inflammation is the murine epididymal and human visceral adipose tissue. The current paradigm is that obesity activates multiple proinflammatory immune cell types in adipose tissue, including adipose-tissue macrophages (ATMs), T Helper 1 (Th1) T cells, and natural killer (NK) cells, while concomitantly suppressing anti-inflammatory immune cells such as T Helper 2 (Th2) T cells and regulatory T cells (Tregs). A key feature of the current paradigm is that obesity induces the anti-inflammatory M2 ATMs in lean adipose tissue to polarize into proinflammatory M1 ATMs. However, recent single-cell transcriptomics studies suggest that the story is much more complex. Here we describe the single-cell genomics technologies that have been developed recently and the emerging results from studies using these technologies. While further studies are needed, it is clear that ATMs are highly heterogeneous. Moreover, while a variety of ATM clusters with quite distinct features have been found to be expanded by obesity, none truly resemble classical M1 ATMs. It is likely that single-cell transcriptomics technology will further revolutionize the field, thereby promoting our understanding of ATMs, adipose-tissue inflammation, and insulin resistance and accelerating the development of therapies for type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Humans , Mice , Animals , Insulin Resistance/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Adipose Tissue/metabolism , Macrophages , Obesity/metabolism , Inflammation/metabolism , Anti-Inflammatory Agents/metabolism , Mice, Inbred C57BL
3.
Mol Cells ; 47(2): 100032, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38369260
4.
Mol Cells ; 47(3): 100007, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38238205

ABSTRACT

Recent evidence establishes a pivotal role for obesity-induced inflammation in precipitating insulin resistance and type-2 diabetes. Central to this process is the proinflammatory M1 adipose-tissue macrophages (ATMs) in epididymal white adipose tissue (eWAT). Notably, natural killer (NK) cells are a crucial regulator of ATMs since their cytokines induce ATM recruitment and M1 polarization. The importance of NK cells is shown by the strong increase in NK-cell numbers in eWAT, and by studies showing that removing and expanding NK cells respectively improve and worsen obesity-induced insulin resistance. It has been suggested that NK cells are activated by unknown ligands on obesity-stressed adipocytes that bind to NKp46 (encoded by Ncr1), which is an activating NK-cell receptor. This was supported by a study showing that NKp46-knockout mice have improved obesity-induced inflammation/insulin resistance. We therefore planned to use the NKp46-knockout mice to further elucidate the molecular mechanism by which NKp46 mediates eWAT NK-cell activation in obesity. We confirmed that obesity increased eWAT NKp46+ NK-cell numbers and NKp46 expression in wild-type mice and that NKp46-knockout ablated these responses. Unexpectedly, however, NKp46-knockout mice demonstrated insulin resistance similar to wild-type mice, as shown by fasting blood glucose/insulin levels and glucose/insulin tolerance tests. Obesity-induced increases in eWAT ATM numbers and proinflammatory gene expression were also similar. Thus, contrary to previously published results, NKp46 does not regulate obesity-induced insulin resistance. It is therefore unclear whether NKp46 participates in the development of obesity-induced inflammation and insulin resistance. This should be considered when elucidating the obesity-mediated molecular mechanisms that activate NK cells.


Subject(s)
Insulin Resistance , Animals , Mice , Inflammation/metabolism , Insulin , Killer Cells, Natural , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Receptors, Natural Killer Cell
5.
Biomater Res ; 27(1): 82, 2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37644502

ABSTRACT

BACKGROUND: Human omentum-derived mesenchymal stem cells (hO-MSCs) possess great potential to differentiate into multiple lineages and have self-renewal capacity, allowing them to be utilized as patient-specific cell-based therapeutics. Although the use of various stem cell-derived ß-cells has been proposed as a novel approach for treating diabetes mellitus, developing an efficient method to establish highly functional ß-cells remains challenging. METHODS: We aimed to develop a novel cell culture platform that utilizes a fibroblast growth factor 2 (FGF2)-immobilized matrix to regulate the adhesion and differentiation of hO-MSCs into insulin-producing ß-cells via cell-matrix/cell-cell interactions. In our study, we evaluated the in vitro differentiation potential of hO-MSCs cultured on an FGF2-immobilized matrix and a round-bottom plate (RBP). Further, the in vivo therapeutic efficacy of the ß-cells transplanted into kidney capsules was evaluated using animal models with streptozotocin (STZ)-induced diabetes. RESULTS: Our findings demonstrated that cells cultured on an FGF2-immobilized matrix could self-organize into insulin-producing ß-cell progenitors, as evident from the upregulation of pancreatic ß-cell-specific markers (PDX-1, Insulin, and Glut-2). Moreover, we observed significant upregulation of heparan sulfate proteoglycan, gap junction proteins (Cx36 and Cx43), and cell adhesion molecules (E-cadherin and Ncam1) in cells cultured on the FGF2-immobilized matrix. In addition, in vivo transplantation of differentiated ß-cells into animal models of STZ-induced diabetes revealed their survival and engraftment as well as glucose-sensitive production of insulin within the host microenvironment, at over 4 weeks after transplantation. CONCLUSIONS: Our findings suggest that the FGF2-immobilized matrix can support initial cell adhesion, maturation, and glucose-stimulated insulin secretion within the host microenvironment. Such a cell culture platform can offer novel strategies to obtain functional pancreatic ß-cells from patient-specific cell sources, ultimately enabling better treatment for diabetes mellitus.

6.
Nat Metab ; 4(2): 155-156, 2022 02.
Article in English | MEDLINE | ID: mdl-35169326
7.
Cell Death Differ ; 29(7): 1423-1432, 2022 07.
Article in English | MEDLINE | ID: mdl-35017647

ABSTRACT

Apoptotic cells are rapidly engulfed and removed by phagocytes after displaying cell surface eat-me signals. Among many phospholipids, only phosphatidylserine (PS) is known to act as an eat-me signal on apoptotic cells. Using unbiased proteomics, we identified externalized phosphatidylinositides (PIPs) as apoptotic eat-me signals recognized by CD14+ phagocytes. Exofacial PIPs on the surfaces of early and late-apoptotic cells were observed in patches and blebs using anti-PI(3,4,5)P3 antibody, AKT- and PLCδ PH-domains, and CD14 protein. Phagocytosis of apoptotic cells was blocked either by masking exofacial PIPs or by CD14 knockout in phagocytes. We further confirmed that exofacial PIP+ thymocytes increased dramatically after in vivo irradiation and that exofacial PIP+ cells represented more significant populations in tissues of Cd14-/- than WT mice, especially after induction of apoptosis. Our findings reveal exofacial PIPs to be previously unknown cell death signals recognized by CD14+ phagocytes.


Subject(s)
Phagocytosis , Signal Transduction , Animals , Apoptosis/physiology , Mice , Phagocytes/metabolism , Phagocytosis/physiology , Phosphatidylserines/metabolism , Signal Transduction/physiology
8.
Ann Pediatr Endocrinol Metab ; 26(1): 1-13, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33819954

ABSTRACT

The prevalence of obesity has increased alarmingly both worldwide and in Korea. This has also dramatically increased the prevalence of chronic obesity-associated diseases, including type 2 diabetes (T2D). Extensive studies on the molecular etiology of T2D have revealed several potential mechanisms by which obesity induces the development of insulin resistance and T2D. One of these is low-grade chronic inflammation. Studies hinting at the existence of this phenomenon were first published about 30 years ago. Ten years later, several seminal papers confirmed its existence, which then led to a rapid and massive escalation of research in this field. Today, the notion that obesity-induced inflammation mediates T2D is now well-accepted. This paper will review the key developments in this field, including the discovery that obesity-induced inflammation and insulin resistance is mainly regulated by adipose tissue-resident immune cells, particularly those in visceral adipose tissue. This review further details the research areas, including (1) the obesity-related factors that induce adipose tissue macrophage (ATM) inflammation, (2) the precise effector functions by which adipose tissue immune cells promote insulin resistance, (3) whether there are early immunological events that have an outsize effect on later events and could be targeted to arrest the development of insulin resistance, (4) the roles played by nonimmunological functions of ATMs and other immune cells, and (5) whether there are noncanonical immune responses to obesity (i.e., immune responses that are unique to obesity and cannot be detected by following the discoveries in the classical immunity field).

9.
Front Immunol ; 10: 944, 2019.
Article in English | MEDLINE | ID: mdl-31134063

ABSTRACT

Metabolic reprogramming during macrophage polarization supports the effector functions of these cells in health and disease. Here, we demonstrate that pyruvate dehydrogenase kinase (PDK), which inhibits the pyruvate dehydrogenase-mediated conversion of cytosolic pyruvate to mitochondrial acetyl-CoA, functions as a metabolic checkpoint in M1 macrophages. Polarization was not prevented by PDK2 or PDK4 deletion but was fully prevented by the combined deletion of PDK2 and PDK4; this lack of polarization was correlated with improved mitochondrial respiration and rewiring of metabolic breaks that are characterized by increased glycolytic intermediates and reduced metabolites in the TCA cycle. Genetic deletion or pharmacological inhibition of PDK2/4 prevents polarization of macrophages to the M1 phenotype in response to inflammatory stimuli (lipopolysaccharide plus IFN-γ). Transplantation of PDK2/4-deficient bone marrow into irradiated wild-type mice to produce mice with PDK2/4-deficient myeloid cells prevented M1 polarization, reduced obesity-associated insulin resistance, and ameliorated adipose tissue inflammation. A novel, pharmacological PDK inhibitor, KPLH1130, improved high-fat diet-induced insulin resistance; this was correlated with a reduction in the levels of pro-inflammatory markers and improved mitochondrial function. These studies identify PDK2/4 as a metabolic checkpoint for M1 phenotype polarization of macrophages, which could potentially be exploited as a novel therapeutic target for obesity-associated metabolic disorders and other inflammatory conditions.


Subject(s)
Macrophage Activation/immunology , Macrophages/immunology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/immunology , Pyruvate Dehydrogenase Complex/immunology , Acetyl Coenzyme A/immunology , Acetyl Coenzyme A/metabolism , Animals , Cytosol/immunology , Cytosol/metabolism , Diet, High-Fat/adverse effects , Insulin Resistance/genetics , Insulin Resistance/immunology , Macrophages/classification , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/immunology , Mitochondria/metabolism , Obesity/etiology , Obesity/genetics , Obesity/immunology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/deficiency , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Pyruvate Dehydrogenase Complex/metabolism , Pyruvic Acid/immunology , Pyruvic Acid/metabolism
10.
Front Immunol ; 9: 696, 2018.
Article in English | MEDLINE | ID: mdl-29731750

ABSTRACT

Obesity-induced chronic low-grade inflammation, in particular in adipose tissue, contributes to the development of insulin resistance and type 2 diabetes. However, the mechanism by which obesity induces adipose tissue inflammation has not been completely elucidated. Recent studies suggest that alteration of the nuclear lamina is associated with age-associated chronic inflammation in humans and fly. These findings led us to investigate whether the nuclear lamina regulates obesity-mediated chronic inflammation. In this study, we show that lamin A/C mediates inflammation in macrophages. The gene and protein expression levels of lamin A/C are significantly increased in epididymal adipose tissues from obese rodent models and omental fat from obese human subjects compared to their lean controls. Flow cytometry and gene expression analyses reveal that the protein and gene expression levels of lamin A/C are increased in adipose tissue macrophages (ATMs) by obesity. We further show that ectopic overexpression of lamin A/C in macrophages spontaneously activates NF-κB, and increases the gene expression levels of proinflammatory genes, such as Il6, Tnf, Ccl2, and Nos2. Conversely, deletion of lamin A/C in macrophages reduces LPS-induced expression of these proinflammatory genes. Importantly, we find that myeloid cell-specific lamin A/C deficiency ameliorates obesity-induced insulin resistance and adipose tissue inflammation. Thus, our data suggest that lamin A/C mediates the activation of ATM inflammation by regulating NF-κB, thereby contributing to the development of obesity-induced insulin resistance.


Subject(s)
Adipose Tissue, White/metabolism , Insulin Resistance , Lamin Type A/metabolism , Macrophages/metabolism , Obesity/metabolism , Animals , Gene Expression , HEK293 Cells , HeLa Cells , Humans , Inflammation/metabolism , Lamin Type A/genetics , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism
11.
Diabetes Metab J ; 41(4): 229-250, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28537058

ABSTRACT

The notion that obesity-induced inflammation mediates the development of insulin resistance in animal models and humans has been gaining strong support. It has also been shown that immune cells in local tissues, in particular in visceral adipose tissue, play a major role in the regulation of obesity-induced inflammation. Specifically, obesity increases the numbers and activation of proinflammatory immune cells, including M1 macrophages, neutrophils, Th1 CD4 T cells, and CD8 T cells, while simultaneously suppressing anti-inflammatory cells such as M2 macrophages, CD4 regulatory T cells, regulatory B cells, and eosinophils. Recently, however, new cell types have been shown to participate in the development of obesity-induced inflammation and insulin resistance. Some of these cell types also appear to regulate obesity. These cells are natural killer (NK) cells and innate lymphoid cells (ILCs), which are closely related, and invariant natural killer T (iNKT) cells. It should be noted that, although iNKT cells resemble NK cells in name, they are actually a completely different cell type in terms of their development and functions in immunity and metabolism. In this review, we will focus on the roles that these relatively new players in the metabolism field play in obesity-induced insulin resistance and the regulation of obesity.

12.
Cell Metab ; 23(4): 685-98, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27050305

ABSTRACT

Obesity-induced inflammation mediated by immune cells in adipose tissue appears to participate in the pathogenesis of insulin resistance. We show that natural killer (NK) cells in adipose tissue play an important role. High-fat diet (HFD) increases NK cell numbers and the production of proinflammatory cytokines, notably TNFα, in epididymal, but not subcutaneous, fat depots. When NK cells were depleted either with neutralizing antibodies or genetic ablation in E4bp4(+/-) mice, obesity-induced insulin resistance improved in parallel with decreases in both adipose tissue macrophage (ATM) numbers, and ATM and adipose tissue inflammation. Conversely, expansion of NK cells following IL-15 administration or reconstitution of NK cells into E4bp4(-/-) mice increased both ATM numbers and adipose tissue inflammation and exacerbated HFD-induced insulin resistance. These results indicate that adipose NK cells control ATMs as an upstream regulator potentially by producing proinflammatory mediators, including TNFα, and thereby contribute to the development of obesity-induced insulin resistance.


Subject(s)
Adipose Tissue/pathology , Inflammation/complications , Insulin Resistance , Killer Cells, Natural/pathology , Macrophages/pathology , Obesity/complications , Adipose Tissue/immunology , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Inflammation/immunology , Inflammation/pathology , Killer Cells, Natural/immunology , Macrophages/immunology , Mice, Inbred C57BL , Obesity/immunology , Obesity/pathology
13.
J Biol Chem ; 289(28): 19769-77, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24867955

ABSTRACT

Complementary surfaces are buried when peptide hormones, growth factors, or cytokines bind and activate cellular receptors. Although these extended surfaces provide high affinity and specificity to the interactions, they also present great challenges to the design of small molecules that might either mimic or antagonize the process. We show that the insulin receptor (IR) and downstream signals can be activated by targeting a site outside of its ligand-binding domain. A 24-residue peptide having the IR transmembrane (TM) domain sequence activates IR, but not related growth factor receptors, through specific interactions with the receptor TM domain. Like insulin-dependent activation, IR-TM requires that IR have a competent ATP-binding site and kinase activation loop. IR-TM also activates mutated receptors from patients with severe insulin resistance, which do not respond to insulin. These results show that IR can be activated through a pathway that bypasses its canonical ligand-binding domain.


Subject(s)
Insulin Resistance , Receptor, Insulin/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Mice , Mutation , NIH 3T3 Cells , Protein Structure, Secondary , Protein Structure, Tertiary , Receptor, Insulin/genetics
14.
Biochim Biophys Acta ; 1842(3): 446-62, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23707515

ABSTRACT

There is increasing evidence showing that inflammation is an important pathogenic mediator of the development of obesity-induced insulin resistance. It is now generally accepted that tissue-resident immune cells play a major role in the regulation of this obesity-induced inflammation. The roles that adipose tissue (AT)-resident immune cells play have been particularly extensively studied. AT contains most types of immune cells and obesity increases their numbers and activation levels, particularly in AT macrophages (ATMs). Other pro-inflammatory cells found in AT include neutrophils, Th1 CD4 T cells, CD8 T cells, B cells, DCs, and mast cells. However, AT also contains anti-inflammatory cells that counter the pro-inflammatory immune cells that are responsible for the obesity-induced inflammation in this tissue. These anti-inflammatory cells include regulatory CD4 T cells (Tregs), Th2 CD4 T cells, and eosinophils. Hence, AT inflammation is shaped by the regulation of pro- and anti-inflammatory immune cell homeostasis, and obesity skews this balance towards a more pro-inflammatory status. Recent genetic studies revealed several molecules that participate in the development of obesity-induced inflammation and insulin resistance. In this review, the cellular and molecular players that participate in the regulation of obesity-induced inflammation and insulin resistance are discussed, with particular attention being placed on the roles of the cellular players in these pathogeneses. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.


Subject(s)
Adipose Tissue/immunology , Inflammation/metabolism , Insulin Resistance/genetics , Obesity/immunology , Adipose Tissue/metabolism , Adipose Tissue/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Humans , Inflammation/immunology , Inflammation/pathology , Insulin Resistance/immunology , Kruppel-Like Factor 4 , Macrophages/cytology , Macrophages/immunology , Neutrophils/immunology , Neutrophils/metabolism , Obesity/metabolism , Obesity/pathology , Th1 Cells/immunology , Th1 Cells/metabolism
15.
PLoS One ; 8(12): e82847, 2013.
Article in English | MEDLINE | ID: mdl-24376593

ABSTRACT

It is increasingly accepted that chronic inflammation participates in obesity-induced insulin resistance and type 2 diabetes (T2D). Salicylates and thiazolidinediones (TZDs) both have anti-inflammatory and anti-hyperglycemic properties. The present study compared the effects of these drugs on obesity-induced inflammation in adipose tissue (AT) and AT macrophages (ATMs), as well as the metabolic and immunological phenotypes of the animal models. Both drugs improved high fat diet (HFD)-induced insulin resistance. However, salicylates did not affect AT and ATM inflammation, whereas Pioglitazone improved these parameters. Interestingly, HFD and the drug treatments all modulated systemic inflammation as assessed by changes in circulating immune cell numbers and activation states. HFD increased the numbers of circulating white blood cells, neutrophils, and a pro-inflammatory monocyte subpopulation (Ly6C(hi)), whereas salicylates and Pioglitazone normalized these cell numbers. The drug treatments also decreased circulating lymphocyte numbers. These data suggest that obesity induces systemic inflammation by regulating circulating immune cell phenotypes and that anti-diabetic interventions suppress systemic inflammation by normalizing circulating immune phenotypes.


Subject(s)
Adipose Tissue/pathology , Diet, High-Fat , Inflammation/drug therapy , Inflammation/pathology , Salicylates/therapeutic use , Thiazolidinediones/therapeutic use , Adipose Tissue/drug effects , Animals , Cell Count , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation/drug effects , Inflammation/genetics , Insulin Resistance , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Multigene Family , Obesity/pathology , Oligonucleotide Array Sequence Analysis , Pioglitazone , Salicylates/pharmacology , Thiazolidinediones/pharmacology
16.
Diabetes ; 62(11): 3718-26, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23884883

ABSTRACT

Metabolic inflammation may contribute to the pathogenesis of obesity and its comorbidities, including type 2 diabetes and cardiovascular disease. Previously, we showed that the actin-binding protein profilin-1 (pfn) plays a role in atherogenesis because pfn heterozygote mice (PfnHet) exhibited a significant reduction in atherosclerotic lesion burden and vascular inflammation. In the current study, we tested whether pfn haploinsufficiency would also limit diet-induced adipose tissue inflammation and insulin resistance (IR). First, we found that a high-fat diet (HFD) upregulated pfn expression in epididymal and subcutaneous white adipose tissue (WAT) but not in the liver or muscle of C57BL/6 mice compared with normal chow. Pfn expression in WAT correlated with F4/80, an established marker for mature macrophages. Of note, HFD elevated pfn protein levels in both stromal vascular cells and adipocytes of WAT. We also found that PfnHet were significantly protected from HFD-induced glucose intolerance observed in pfn wild-type mice. With HFD, PfnHet displayed blunted expression of systemic and WAT proinflammatory cytokines and decreased accumulation of adipose tissue macrophages, which were also preferentially biased toward an M2-like phenotype; this correlated with preserved frequency of regulatory T cells. Taken together, the findings indicate that pfn haploinsufficiency protects against diet-induced IR and inflammation by modulating WAT immune homeostasis.


Subject(s)
Adipose Tissue, White/immunology , Glucose Intolerance/genetics , Haploinsufficiency , Inflammation/immunology , Profilins/genetics , Subcutaneous Fat/immunology , Adipose Tissue, White/pathology , Animals , Antigens, Differentiation/biosynthesis , Diet, High-Fat , Homeostasis , Insulin Resistance/physiology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Profilins/biosynthesis , STAT3 Transcription Factor/metabolism , Subcutaneous Fat/metabolism
17.
Arch Pharm Res ; 36(2): 208-22, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23397293

ABSTRACT

It has been increasingly accepted that chronic subacute inflammation plays an important role in the development of insulin resistance and type 2 diabetes in animals and humans. Particularly supporting this is that suppression of systemic inflammation in type 2 diabetes improves glycemic control; this also points to a new potential therapeutic target for the treatment of type 2 diabetes. Recent studies strongly suggest that obesity-induced inflammation is mainly mediated by tissue resident immune cells, with particular attention being focused on adipose tissue macrophages (ATMs). This review delineates the current progress made in understanding obesity-induced inflammation and the roles ATMs play in this process.


Subject(s)
Adipose Tissue/pathology , Diabetes Mellitus, Type 2/pathology , Insulin Resistance/physiology , Macrophages/pathology , Obesity/pathology , Adipose Tissue/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Humans , Inflammation/etiology , Inflammation/pathology , Inflammation/physiopathology , Macrophages/metabolism , Obesity/metabolism
18.
Invest Ophthalmol Vis Sci ; 53(13): 8424-32, 2012 Dec 19.
Article in English | MEDLINE | ID: mdl-23197686

ABSTRACT

PURPOSE: To correlate changes between VEGF expression with systemic and retinal oxidative stress and inflammation in rodent models of obesity induced insulin resistance and diabetes. METHODS: Retinal VEGF mRNA and protein levels were assessed by RT-PCR and VEGF ELISA, respectively. Urinary 8-hydroxydeoxyguanosine (8-OHdG), blood levels of C-reactive protein (CRP), malondialdehyde (MDA), and CD11b/c positive cell ratio were used as systemic inflammatory markers. Retinal expression of Nox2, Nox4, and p47phox mRNA levels were measured as oxidative stress markers. TNF-α, inter-cellular adhesion molecule-1 (ICAM-1), IL1ß, and activation of nuclear factor κB (NF-κB) were used as retinal inflammatory markers. RESULTS: Retinal VEGF mRNA and protein expression increased in Zucker diabetic fatty (ZDF(fa/fa)) rats and streptozotosin (STZ) induced diabetic Sprague-Dawley rats, after two months of disease, but not in Zucker fatty (ZF) rats. Systemic markers of oxidative stress and inflammation were elevated in insulin resistant and diabetic rats. Some oxidative stress and inflammatory markers (TNF-α, IL-6, ICAM-1, and IL1-ß) were upregulated in the retina of ZDF(fa/fa) and STZ diabetic rats after 4 months of disease. In contrast, activation of NF-κB in the retina was observed in high fat fed nondiabetic and diabetic cis-NF-κB(EGFP) mice, ZF, ZDF(fa/fa), and STZ-induced diabetic rats. CONCLUSIONS: Only persistent hyperglycemia and diabetes increased retinal VEGF expression. Some markers of inflammation and oxidative stress were elevated in the retina and systemic circulation of obese and insulin resistant rodents with and without diabetes. Induction of VEGF and its associated retinal pathologies by diabetes requires chronic hyperglycemia and factors in addition to inflammation and oxidative stress.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Insulin Resistance/physiology , Oxidative Stress/physiology , Retina/metabolism , Stress, Physiological/physiology , Vascular Endothelial Growth Factor A/metabolism , 8-Hydroxy-2'-Deoxyguanosine , Animals , Biomarkers/metabolism , C-Reactive Protein/metabolism , CD11b Antigen/metabolism , CD11c Antigen/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/urine , Enzyme-Linked Immunosorbent Assay , Inflammation/metabolism , Male , Malondialdehyde/blood , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Obesity/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Rats, Zucker , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics
19.
Arterioscler Thromb Vasc Biol ; 32(8): 1771-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22815343

ABSTRACT

Obesity and its comorbidities, including type 2 diabetes mellitus and cardiovascular disease, are associated with a state of chronic low-grade inflammation that can be detected both systemically and within specific tissues. Areas of active investigation focus on the molecular bases of metabolic inflammation and potential pathogenic roles in insulin resistance, diabetes, and cardiovascular disease. An increased accumulation of macrophages occurring in obese adipose tissue has emerged as a key process in metabolic inflammation. Recent studies have also begun to unravel the heterogeneity of adipose tissue macrophages, and their physical and functional interactions with adipocytes, endothelial cells, and other immune cells within the adipose tissue microenvironment. Translating the information gathered from experimental models of insulin resistance and diabetes into meaningful therapeutic interventions is a tantalizing goal with long-term global health implications. In this context, ongoing clinical studies are testing the effects of targeting inflammation systemically on metabolic and cardiovascular outcomes.


Subject(s)
Inflammation/complications , Insulin Resistance , Metabolic Syndrome/complications , Adipose Tissue, White/physiology , Animals , Anti-Inflammatory Agents/therapeutic use , Chronic Disease , Humans , Leukocytes/physiology , Macrophages/physiology , Obesity/complications , Weight Loss
20.
Nature ; 486(7404): 549-53, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22722857

ABSTRACT

Obesity and type-2 diabetes have increased markedly over the past few decades, in parallel. One of the major links between these two disorders is chronic, low-grade inflammation. Prolonged nutrient excess promotes the accumulation and activation of leukocytes in visceral adipose tissue (VAT) and ultimately other tissues, leading to metabolic abnormalities such as insulin resistance, type-2 diabetes and fatty-liver disease. Although invasion of VAT by pro-inflammatory macrophages is considered to be a key event driving adipose-tissue inflammation and insulin resistance, little is known about the roles of other immune system cell types in these processes. A unique population of VAT-resident regulatory T (Treg) cells was recently implicated in control of the inflammatory state of adipose tissue and, thereby, insulin sensitivity. Here we identify peroxisome proliferator-activated receptor (PPAR)-γ, the 'master regulator' of adipocyte differentiation, as a crucial molecular orchestrator of VAT Treg cell accumulation, phenotype and function. Unexpectedly, PPAR-γ expression by VAT Treg cells was necessary for complete restoration of insulin sensitivity in obese mice by the thiazolidinedione drug pioglitazone. These findings suggest a previously unknown cellular mechanism for this important class of thiazolidinedione drugs, and provide proof-of-principle that discrete populations of Treg cells with unique functions can be precisely targeted to therapeutic ends.


Subject(s)
Adipose Tissue/cytology , PPAR gamma/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Adipose Tissue/immunology , Adipose Tissue/pathology , Animals , Cell Differentiation , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Epididymis/cytology , Epididymis/immunology , Forkhead Transcription Factors/metabolism , Gene Expression , Hypoglycemic Agents/pharmacology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Insulin Resistance/physiology , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Obesity/pathology , Phenotype , Pioglitazone , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes, Regulatory/drug effects , Thiazolidinediones/pharmacology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...