Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Arch Biochem Biophys ; 711: 109029, 2021 10 30.
Article in English | MEDLINE | ID: mdl-34517011

ABSTRACT

Because of the critical roles of Toll-like receptors (TLRs) and receptor for advanced glycation end-products (RAGE) in the pathophysiology of various acute and chronic inflammatory diseases, continuous efforts have been made to discover novel therapeutic inhibitors of TLRs and RAGE to treat inflammatory disorders. A recent study by our group has demonstrated that trimebutine, a spasmolytic drug, suppresses the high mobility group box 1‒RAGE signaling that is associated with triggering proinflammatory signaling pathways in macrophages. Our present work showed that trimebutine suppresses interleukin-6 (IL-6) production in lipopolysaccharide (LPS, a stimulant of TLR4)-stimulated macrophages of RAGE-knockout mice. In addition, trimebutine suppresses the LPS-induced production of various proinflammatory cytokines and chemokines in mouse macrophage-like RAW264.7 cells. Importantly, trimebutine suppresses IL-6 production induced by TLR2-and TLR7/8/9 stimulants. Furthermore, trimebutine greatly reduces mortality in a mouse model of LPS-induced sepsis. Studies exploring the action mechanism of trimebutine revealed that it inhibits the LPS-induced activation of IL-1 receptor-associated kinase 1 (IRAK1), and the subsequent activations of extracellular signal-related kinase 1/2 (ERK1/2), c-Jun N-terminal kinase (JNK), and nuclear factor-κB (NF-κB). These findings suggest that trimebutine exerts anti-inflammatory effects on TLR signaling by downregulating IRAK1‒ERK1/2‒JNK pathway and NF-κB activity, thereby indicating the therapeutic potential of trimebutine in inflammatory diseases. Therefore, trimebutine can be a novel anti-inflammatory drug-repositioning candidate and may provide an important scaffold for designing more effective dual anti-inflammatory drugs that target TLR/RAGE signaling.


Subject(s)
Anti-Inflammatory Agents/pharmacology , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Toll-Like Receptors/metabolism , Trimebutine/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Chemokines/metabolism , Female , Interleukin-6/metabolism , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells , Receptor for Advanced Glycation End Products/deficiency , Receptor for Advanced Glycation End Products/genetics , Sepsis/chemically induced , Sepsis/drug therapy , Trimebutine/therapeutic use
2.
Elife ; 102021 02 16.
Article in English | MEDLINE | ID: mdl-33588989

ABSTRACT

Influenza virus has a high mutation rate, such that within one host different viral variants can emerge. Evidence suggests that influenza virus variants are more prevalent in pregnant and/or obese individuals due to their impaired interferon response. We have recently shown that the non-allergic, paucigranulocytic subtype of asthma is associated with impaired type I interferon production. Here, we seek to address if this is associated with an increased emergence of influenza virus variants. Compared to controls, mice with paucigranulocytic asthma had increased disease severity and an increased emergence of influenza virus variants. Specifically, PB1 mutations exclusively detected in asthmatic mice were associated with increased polymerase activity. Furthermore, asthmatic host-derived virus led to increased disease severity in wild-type mice. Taken together, these data suggest that at least a subset of patients with asthma may be more susceptible to severe influenza and may be a possible source of new influenza virus variants.


Subject(s)
Asthma/virology , Influenza A Virus, H1N1 Subtype/pathogenicity , Orthomyxoviridae Infections/virology , Animals , Female , Host-Pathogen Interactions , Male , Mice , Mice, Inbred C57BL , Receptor for Advanced Glycation End Products/deficiency
3.
Angiogenesis ; 24(1): 47-55, 2021 02.
Article in English | MEDLINE | ID: mdl-32989644

ABSTRACT

Peripheral arterial disease occurs more frequently and has a worse prognosis in patients with chronic kidney disease (CKD). The receptor for advanced glycation end products (RAGE) is involved in multiple aspects of uremia-associated vasculopathy. Previous data suggest that the RAGE pathway may promote soluble fms-like tyrosine kinase 1 (sFlt1) production, an anti-angiogenic molecule. Thus, we tested the hypothesis that the deletion of AgeR would decrease sFlt1 production and improve post-ischemic revascularization in uremic condition. We used a well-established CKD model (5/6 nephrectomy) in WT and AgeR-/- C57/Bl6 mice. Hindlimb ischemia was induced by femoral artery ligation. Revascularization was evaluated by complementary approaches: ischemic limb retraction, LASCA imagery, and capillary density. The production of sFlt1 was assessed at both RNA and protein levels. After hindlimb ischemia, uremic mice showed slower functional recovery (p < 0.01), decreased reperfusion (p < 0.01), lower capillary density (p = 0.02), and increased circulating sFlt1 levels (p = 0.03). AgeR deletion restored post-ischemic angiogenesis and was protective from sFlt1 increase in uremic mice. These findings show the main role of RAGE in post-ischemic angiogenesis impairment associated with CKD. RAGE may represent a key target for building new therapeutic approaches to improve the outcome of CKD patients with PAD.


Subject(s)
Gene Deletion , Ischemia/complications , Neovascularization, Physiologic , Receptor for Advanced Glycation End Products/deficiency , Uremia/complications , Vascular Endothelial Growth Factor Receptor-1/biosynthesis , Animals , Biomarkers/blood , Cell Line , Humans , Ligands , Male , Mice, Inbred C57BL , RNA/metabolism , Receptor for Advanced Glycation End Products/metabolism , Solubility , Up-Regulation
4.
Int J Mol Sci ; 21(19)2020 Oct 01.
Article in English | MEDLINE | ID: mdl-33019603

ABSTRACT

We addressed the involvement of the receptor for advanced glycation end products (RAGE) in the impairment of the cellular cholesterol efflux elicited by glycated albumin. Albumin was isolated from type 1 (DM1) and type 2 (DM2) diabetes mellitus (HbA1c > 9%) and non-DM subjects (C). Moreover, albumin was glycated in vitro (AGE-albumin). Macrophages from Ager null and wild-type (WT) mice, or THP-1 transfected with siRNA-AGER, were treated with C, DM1, DM2, non-glycated or AGE-albumin. The cholesterol efflux was reduced in WT cells exposed to DM1 or DM2 albumin as compared to C, and the intracellular lipid content was increased. These events were not observed in Ager null cells, in which the cholesterol efflux and lipid staining were, respectively, higher and lower when compared to WT cells. In WT, Ager, Nox4 and Nfkb1, mRNA increased and Scd1 and Abcg1 diminished after treatment with DM1 and DM2 albumin. In Ager null cells treated with DM-albumin, Nox4, Scd1 and Nfkb1 were reduced and Jak2 and Abcg1 increased. In AGER-silenced THP-1, NOX4 and SCD1 mRNA were reduced and JAK2 and ABCG1 were increased even after treatment with AGE or DM-albumin. RAGE mediates the deleterious effects of AGE-albumin in macrophage cholesterol efflux.


Subject(s)
Cholesterol, HDL/blood , Cholesterol, LDL/blood , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Macrophages/metabolism , Receptor for Advanced Glycation End Products/genetics , Adult , Animals , Case-Control Studies , Cell Line , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Glycated Hemoglobin/genetics , Glycated Hemoglobin/metabolism , Glycation End Products, Advanced/blood , Glycation End Products, Advanced/pharmacology , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Receptor for Advanced Glycation End Products/deficiency , Receptor for Advanced Glycation End Products/metabolism , Serum Albumin, Human/metabolism , Serum Albumin, Human/pharmacology , THP-1 Cells , Triglycerides/blood
5.
J Vet Diagn Invest ; 32(5): 664-674, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32715975

ABSTRACT

Innate immunity plays a central role in the pathogenesis of chronic inflammatory enteropathies (CIE) in dogs, and further evaluation of the innate immune receptor for advanced glycation end-products (RAGE) is warranted. We measured serum concentrations of decoy receptor soluble RAGE (sRAGE) in 102 dogs diagnosed with CIE, and evaluated relationships with clinical disease severity, histologic lesion severity, concentrations of serum C-reactive protein (CRP), and serum and fecal calprotectin, S100A12, and alpha1-proteinase inhibitor (α1PI). Serum sRAGE levels were not associated with clinical disease activity, serum CRP, serum and fecal α1PI, calprotectin, or S100A12 concentrations. Microscopic lesions in the duodenum were more severe in dogs with serum sRAGE concentration ≤ 340 ng/L (p = 0.013). Serum sRAGE levels were weakly and inversely correlated with the severity of lymphoplasmacytic infiltration in the gastric antrum and duodenum, and with crypt dilation and the neutrophilic infiltrate in the duodenum, in univariate analysis (all p < 0.05), but none of the correlations remained statistically significant after correction for multiple comparisons. Our study confirms that CIE in dogs is associated with decreased serum sRAGE concentrations, suggesting a dysregulated sRAGE/RAGE axis.


Subject(s)
Blood/metabolism , Dog Diseases/diagnosis , Inflammation/veterinary , Receptor for Advanced Glycation End Products/deficiency , Animals , Biomarkers/blood , Blood Chemical Analysis/veterinary , Chronic Disease/veterinary , Dogs , Female , Inflammation/diagnosis , Male , Receptor for Advanced Glycation End Products/blood , Severity of Illness Index
6.
Brain Res Bull ; 163: 49-56, 2020 10.
Article in English | MEDLINE | ID: mdl-32621862

ABSTRACT

A pivotal role of glutamatergic neurotransmission in the pathophysiology of major depressive disorder (MDD) has been supported in preclinical and clinical studies. Glutamate transporters are responsible for rapid uptake of glutamate to maintain glutamate homeostasis. Down-regulation of glutamate transporters has been reported in MDD patients and animal models. However, the mechanism for stress-induced modulation of glutamate transporter expression is poorly understood. Receptor for advanced glycosylation end products (RAGE), a member of immunoglobulin family, is found expressed widely in brain and play important roles in neuronal development, neurite growth, neurogenesis and neuroinflammation. Our study showed chronic unpredictable stress (CUS) induced depressive-like behaviors and reduced RAGE expression in hippocampus DG, CA1 and CA3 areas. The protein levels of GLT-1, p-CREB and p-p65 decreased in hippocampus DG as well. Knockdown of RAGE expression in hippocampus DG with RAGE shRNA lentivirus particles induced depressive-like behaviors. Meanwhile, the protein and mRNA levels of GLT-1 were significantly decreased as well as phosphorylation of CREB and p65. Neither CUS nor RAGE knockdown altered GLAST protein and mRNA levels. These findings suggested that RAGE/CREB-NF-κB signaling pathway in hippocampus DG involved in modulation of GLT-1 expression, which possibly contributed to the depressive-like behavior induced by CUS.


Subject(s)
Dentate Gyrus/metabolism , Excitatory Amino Acid Transporter 2/biosynthesis , Receptor for Advanced Glycation End Products/deficiency , Signal Transduction/physiology , Stress, Psychological/metabolism , Animals , Chronic Disease , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/genetics , Gene Knockdown Techniques/methods , Male , Rats , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products/genetics , Stress, Psychological/genetics , Stress, Psychological/psychology
7.
Mol Neurobiol ; 57(10): 4143-4155, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32676989

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly progressing motor neuron degenerative disease that is without effective treatment. The receptor for advanced glycation end products (RAGE) is a major component of the innate immune system that has been implicated in ALS pathogenesis. However, the contribution of RAGE signalling to the neuroinflammation that underlies ALS neurodegeneration remains unknown. The present study therefore generated SOD1G93A mice lacking RAGE and compared them with SOD1G93A transgenic ALS mice in respect to disease progression (i.e. body weight, survival and muscle strength), neuroinflammation and denervation markers in the spinal cord and tibialis anterior muscle. We found that complete absence of RAGE signalling exerted a protective effect on SOD1G93A pathology, slowing disease progression and significantly extending survival by ~ 3 weeks and improving motor function (rotarod and grip strength). This was associated with reduced microgliosis, cytokines, innate immune factors (complement, TLRs, inflammasomes), and oxidative stress in the spinal cord, and a reduction of denervation markers in the tibialis anterior muscle. We also documented that RAGE mRNA expression was significantly increased in the spinal cord and muscles of preclinical SOD1 and TDP43 models of ALS, supporting a widespread involvement for RAGE in ALS pathology. In summary, our results indicate that RAGE signalling drives neuroinflammation and contributes to neurodegeneration in ALS and highlights RAGE as a potential immune therapeutic target for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Inflammation/pathology , Receptor for Advanced Glycation End Products/deficiency , Superoxide Dismutase-1/genetics , Animals , Astrocytes/pathology , Biomarkers/metabolism , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Denervation , Disease Models, Animal , Disease Progression , Gene Deletion , Hand Strength , Hindlimb/physiopathology , Macrophages/pathology , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Muscles/innervation , Muscles/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Rotarod Performance Test , Severity of Illness Index , Spinal Cord/pathology , Survival Analysis , Up-Regulation
8.
Int J Mol Sci ; 20(13)2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31266174

ABSTRACT

It is incompletely understood how self-antigens become targets of humoral immunity in antibody-mediated autoimmune diseases. In this context, alarmins are discussed as an important level of regulation. Alarmins are recognized by various receptors, such as receptor for advanced glycation end products (RAGE). As RAGE is upregulated under inflammatory conditions, strongly binds nucleic acids and mediates pro-inflammatory responses upon alarmin recognition, our aim was to examine its contribution to immune complex-mediated autoimmune diseases. This question was addressed employing RAGE-/- animals in murine models of pristane-induced lupus, collagen-induced, and serum-transfer arthritis. Autoantibodies were assessed by enzyme-linked immunosorbent assay, renal disease by quantification of proteinuria and histology, arthritis by scoring joint inflammation. The associated immune status was determined by flow cytometry. In both disease entities, we detected tendentiously decreased autoantibody levels in RAGE-/- mice, however no differences in clinical outcome. In accordance with autoantibody levels, a subgroup of the RAGE-/- animals showed a decrease in plasma cells, and germinal center B cells and an increase in follicular B cells. Based on our results, we suggest that RAGE deficiency alone does not significantly affect antibody-mediated autoimmunity. RAGE may rather exert its effects along with other receptors linking environmental factors to auto-reactive immune responses.


Subject(s)
Arthritis, Experimental/immunology , Autoantibodies/metabolism , Lupus Nephritis/immunology , Receptor for Advanced Glycation End Products/deficiency , Animals , Arthritis, Experimental/genetics , Autoantibodies/blood , B-Lymphocytes/immunology , Collagen/adverse effects , Disease Models, Animal , Germinal Center/immunology , Lupus Nephritis/genetics , Mice , Receptor for Advanced Glycation End Products/immunology , Terpenes/adverse effects
9.
Aging Cell ; 18(2): e12850, 2019 04.
Article in English | MEDLINE | ID: mdl-30794349

ABSTRACT

Pro-aging effects of endogenous advanced glycation end-products (AGEs) have been reported, and there is increasing interest in the pro-inflammatory and -fibrotic effects of their binding to RAGE (the main AGE receptor). The role of dietary AGEs in aging remains ill-defined, but the predominantly renal accumulation of dietary carboxymethyllysine (CML) suggests the kidneys may be particularly affected. We studied the impact of RAGE invalidation and a CML-enriched diet on renal aging. Two-month-old male, wild-type (WT) and RAGE-/- C57Bl/6 mice were fed a control or a CML-enriched diet (200 µg CML/gfood ) for 18 months. Compared to controls, we observed higher CML levels in the kidneys of both CML WT and CML RAGE-/- mice, with a predominantly tubular localization. The CML-rich diet had no significant impact on the studied renal parameters, whereby only a trend to worsening glomerular sclerosis was detected. Irrespective of diet, RAGE-/- mice were significantly protected against nephrosclerosis lesions (hyalinosis, tubular atrophy, fibrosis and glomerular sclerosis) and renal senile apolipoprotein A-II (ApoA-II) amyloidosis (p < 0.001). A positive linear correlation between sclerosis score and ApoA-II amyloidosis score (r = 0.92) was observed. Compared with old WT mice, old RAGE-/- mice exhibited lower expression of inflammation markers and activation of AKT, and greater expression of Sod2 and SIRT1. Overall, nephrosclerosis lesions and senile amyloidosis were significantly reduced in RAGE-/- mice, indicating a protective effect of RAGE deletion with respect to renal aging. This could be due to reduced inflammation and oxidative stress in RAGE-/- mice, suggesting RAGE is an important receptor in so-called inflamm-aging.


Subject(s)
Aging/metabolism , Kidney Diseases/metabolism , Receptor for Advanced Glycation End Products/metabolism , Animals , Kidney Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor for Advanced Glycation End Products/deficiency
10.
Sci Rep ; 9(1): 231, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30659203

ABSTRACT

The receptor for advanced glycation end products (RAGE), a cell membrane receptor, recognizes ligands produced by cigarette smoke (CS) and has been implicated in the pathogenesis of COPD. We demonstrate that deletion or pharmacologic inhibition of RAGE prevents development of CS-induced emphysema. To identify molecular pathways by which RAGE mediates smoking related lung injury we performed unbiased gene expression profiling of alveolar macrophages (AM) obtained from RAGE null and C57BL/6 WT mice exposed to CS for one week or four months. Pathway analysis of RNA expression identified a number of genes integral to the pathogenesis of COPD impacted by the absence of RAGE. Altered expression of antioxidant response genes and lung protein 4-HNE immunostaining suggest attenuated oxidative stress in the RAGE null mice despite comparable CS exposure and lung leukocyte burden as the WT mice. Reduced endoplasmic reticulum stress in response to CS exposure also was observed in the AM from RAGE null mice. These findings provide novel insight into the sources of oxidative stress, macrophage activation, and the pathogenesis of lung disease due to CS exposure.


Subject(s)
Cigarette Smoking/adverse effects , Emphysema/physiopathology , Lung/pathology , Macrophage Activation , Macrophages, Alveolar/immunology , Oxidative Stress , Receptor for Advanced Glycation End Products/metabolism , Animals , Disease Models, Animal , Gene Expression Profiling , Mice, Inbred C57BL , Mice, Knockout , Receptor for Advanced Glycation End Products/deficiency , Smoke/adverse effects
11.
Fukushima J Med Sci ; 65(3): 109-121, 2019.
Article in English | MEDLINE | ID: mdl-31915324

ABSTRACT

An advanced glycation end products (AGE)/a receptor for AGE (RAGE) axis plays a central role in the pathogenesis of diabetic vascular remodeling. This study was conducted to clarify the role of RAGE in nondiabetic atherosclerosis. We used the aortic and coronary atherosclerotic lesions of Watanabe heritable hyperlipidemic (WHHL) rabbits prone to myocardial infarction (WHHLMI) at 1 to 14 months. Immunohistochemistry demonstrated the significant expression of RAGE as early as at 1 month with the stronger expression at 3 and 7 months, which was remarkably diminished at 14 months. RAGE expression was concordant with AGE accumulation. The major original sources of RAGE expression were macrophages and smooth muscle cells in addition to endothelial cells, and RAGE expression was distributed in the areas of phospholipid products, a component of oxidized LDL and nitrotyrosine. The concentrations of serum AGE did not alter significantly with aging. These findings suggested the expression of RAGE was induced by hyperlipidemia and oxidative stress independent of diabetes in WHHLMI rabbits. Additionally, our in vitro study showed that silencing of RAGE tended to attenuate oxidized-LDL-triggered PAI-1 expression in human cultured macrophages, as well as oxidized-LDL-induced tissue factor expression in peritoneal macrophages, suggesting a possible role of RAGE in prothrombogenic molecular regulation. In conclusion, the present study provides in vivo evidence that RAGE plays an integral role in the initiation and progression of nondiabetic atherosclerosis, suggesting that RAGE may be a novel target for treating not only diabetic but also nondiabetic vascular complications.


Subject(s)
Atherosclerosis/metabolism , Receptor for Advanced Glycation End Products/metabolism , Aging/metabolism , Aging/pathology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Atherosclerosis/etiology , Atherosclerosis/pathology , Disease Models, Animal , Female , Gene Knockdown Techniques , Glycation End Products, Advanced/blood , Glycation End Products, Advanced/metabolism , Humans , Hyperlipidemias/complications , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Immunohistochemistry , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Oxidative Stress , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Rabbits , Receptor for Advanced Glycation End Products/deficiency , Receptor for Advanced Glycation End Products/genetics , Tyrosine/analogs & derivatives , Tyrosine/metabolism
12.
Adv Exp Med Biol ; 1074: 185-191, 2018.
Article in English | MEDLINE | ID: mdl-29721943

ABSTRACT

The eye is an immuno-privileged organ. However, certain diseases such as uveitis are intrinsically linked to inflammation. In several retinal degenerative diseases, there is a unique damage at the onset of the disease, but evidence suggests that chronic and low-grade inflammatory processes play an important role in their progression. Studies have identified similar signaling pathways and changes in resident immune cells within the retina among these diseases. Herein, we will discuss some of these studies and propose how understanding this inflammatory response could aid in the development of therapies.


Subject(s)
Diabetic Retinopathy/immunology , Macular Degeneration/immunology , Retinitis Pigmentosa/immunology , Animals , Antigens, Neoplasm/physiology , Cytokines/physiology , Diabetic Retinopathy/pathology , Ependymoglial Cells/immunology , Ependymoglial Cells/pathology , Gliosis/immunology , Gliosis/pathology , Humans , Inflammasomes/physiology , Inflammation , Macular Degeneration/pathology , Mice , Microglia/immunology , Microglia/pathology , Mitogen-Activated Protein Kinases/physiology , Receptor for Advanced Glycation End Products/deficiency , Retina/immunology , Retina/pathology , Retinal Drusen/immunology , Retinal Drusen/pathology , Retinitis Pigmentosa/pathology
13.
J Infect Dis ; 218(5): 791-800, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29329449

ABSTRACT

Background: Lack of receptor for advanced glycation end products (RAGE) ameliorates several infections including Staphylococcus aureus pneumonia. We sought to investigate the role of RAGE in staphylococcal skin infection in mice. Methods: Wild-type (WT) and RAGE deficient (RAGE-/-) mice were subcutaneously inoculated with S. aureus SH1000 strain in abscess-forming dose or necrotic dose. Clinical signs of dermatitis, along with histopathological changes, were compared between the groups. Results: The skin lesion size was smaller in RAGE-/- mice. Infected RAGE-/- mice expressed lower proinflammatory cytokines in local skins compared to control mice. Low dose of bacteria caused more abscess formation in RAGE-/- mice compared to skin necrosis that was more often observed in WT mice. As a result of more abscess formation, the wound healing was prolonged in RAGE-/- mice. Importantly, RAGE-/- mice had lower bacterial loads in the skin than controls, which is correlated with higher local levels of myeloperoxidase before skin infection. In vitro, enhanced phagocytic capacity of neutrophils and macrophages obtained from RAGE-/- mice compared to control mice was observed. Conclusions: RAGE deficiency up-regulates phagocytic capacity of phagocytes, resulting in lower bacterial burden in local skin and milder skin lesions in mice with staphylococcal skin infection.


Subject(s)
Abscess/pathology , Receptor for Advanced Glycation End Products/deficiency , Skin/pathology , Staphylococcal Skin Infections/pathology , Wound Healing , Abscess/genetics , Animals , Bacterial Load , Cytokines/analysis , Disease Models, Animal , Female , Histocytochemistry , Macrophages/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Phagocytosis , Receptor for Advanced Glycation End Products/metabolism , Staphylococcal Skin Infections/genetics
14.
Oxid Med Cell Longev ; 2017: 4082102, 2017.
Article in English | MEDLINE | ID: mdl-29209448

ABSTRACT

Sepsis is a threatening health problem and characterized by microvascular dysfunction. In this study, we verified that LPS caused the downregulation of Sirt1 and the hyperpermeability of endothelial cells. Inhibition of Sirt1 with ex527 or Sirt1 siRNA displayed a higher permeability, while activation of Sirt1 with SRT1720 reversed the LPS-induced hyperpermeability, formation of fiber stress, and disruption of VE-cadherin distribution. In pulmonary microvascular vein endothelial cells isolated from wild-type mice, Sirt1 was attenuated upon LPS, while Sirt1 was preserved in a receptor of advanced glycation end product-knockout mice. The RAGE antibody could also diminish the downregulation and ubiquitination of Sirt1 in LPS-exposed human umbilical vein endothelial cells. An LPS-induced decrease in Sirt1 activity was attenuated by the RAGE antibody and TLR4 inhibitor. In vivo study also demonstrated the attenuating role of Sirt1 and RAGE knockout in LPS-induced increases in dextran leakage of mesenteric venules. Furthermore, activation of Sirt1 prevented LPS-induced decreases in the activity and expression of superoxide dismutase 2, as well as the increases in NADPH oxidase 4 and reactive oxygen species, while inhibition of Sirt1 aggravated the SOD2 decline. It also demonstrated that Sirt1-deacetylated p53 is required for p53 inactivation, which reversed the downregulation of ß-catenin caused by LPS.


Subject(s)
Down-Regulation/drug effects , Lipopolysaccharides/toxicity , Permeability/drug effects , Sirtuin 1/metabolism , Animals , Heterocyclic Compounds, 4 or More Rings/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 4/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products/deficiency , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/immunology , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Superoxide Dismutase/metabolism , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism , Tumor Suppressor Protein p53/metabolism , beta Catenin/metabolism
15.
PLoS One ; 12(9): e0184331, 2017.
Article in English | MEDLINE | ID: mdl-28926576

ABSTRACT

World Trade Center-particulate matter(WTC-PM) exposure and metabolic-risk are associated with WTC-Lung Injury(WTC-LI). The receptor for advanced glycation end-products (RAGE) is most highly expressed in the lung, mediates metabolic risk, and single-nucleotide polymorphisms at the AGER-locus predict forced expiratory volume(FEV). Our objectives were to test the hypotheses that RAGE is a biomarker of WTC-LI in the FDNY-cohort and that loss of RAGE in a murine model would protect against acute PM-induced lung disease. We know from previous work that early intense exposure at the time of the WTC collapse was most predictive of WTC-LI therefore we utilized a murine model of intense acute PM-exposure to determine if loss of RAGE is protective and to identify signaling/cytokine intermediates. This study builds on a continuing effort to identify serum biomarkers that predict the development of WTC-LI. A case-cohort design was used to analyze a focused cohort of male never-smokers with normal pre-9/11 lung function. Odds of developing WTC-LI increased by 1.2, 1.8 and 1.0 in firefighters with soluble RAGE (sRAGE)≥97pg/mL, CRP≥2.4mg/L, and MMP-9≤397ng/mL, respectively, assessed in a multivariate logistic regression model (ROCAUC of 0.72). Wild type(WT) and RAGE-deficient(Ager-/-) mice were exposed to PM or PBS-control by oropharyngeal aspiration. Lung function, airway hyperreactivity, bronchoalveolar lavage, histology, transcription factors and plasma/BAL cytokines were quantified. WT-PM mice had decreased FEV and compliance, and increased airway resistance and methacholine reactivity after 24-hours. Decreased IFN-γ and increased LPA were observed in WT-PM mice; similar findings have been reported for firefighters who eventually develop WTC-LI. In the murine model, lack of RAGE was protective from loss of lung function and airway hyperreactivity and was associated with modulation of MAP kinases. We conclude that in a multivariate adjusted model increased sRAGE is associated with WTC-LI. In our murine model, absence of RAGE mitigated acute deleterious effects of PM and may be a biologically plausible mediator of PM-related lung disease.


Subject(s)
Forced Expiratory Volume/drug effects , Lung Injury/etiology , Lung Injury/physiopathology , Particulate Matter/toxicity , Receptor for Advanced Glycation End Products/metabolism , September 11 Terrorist Attacks , Acute Disease , Adult , Animals , Biomarkers/analysis , Biomarkers/blood , Bronchial Hyperreactivity/etiology , Bronchoalveolar Lavage Fluid/chemistry , Case-Control Studies , Cohort Studies , Disease Models, Animal , Female , Firefighters , Humans , Lung/drug effects , Lung/pathology , Lung/physiology , Lung Injury/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Receptor for Advanced Glycation End Products/deficiency , Receptor for Advanced Glycation End Products/genetics
16.
mBio ; 8(4)2017 08 22.
Article in English | MEDLINE | ID: mdl-28830942

ABSTRACT

For more than a century, diabetic patients have been considered immunosuppressed due to defects in phagocytosis and microbial killing. We confirmed that diabetic mice were hypersusceptible to bacteremia caused by Gram-negative bacteria (GNB), dying at inocula nonlethal to nondiabetic mice. Contrary to the pervasive paradigm that diabetes impedes phagocytic function, the bacterial burden was no greater in diabetic mice despite excess mortality. However, diabetic mice did exhibit dramatically increased levels of proinflammatory cytokines in response to GNB infections, and immunosuppressing these cytokines with dexamethasone restored their resistance to infection, both of which are consistent with excess inflammation. Furthermore, disruption of the receptor for advanced glycation end products (RAGE), which is stimulated by heightened levels of AGEs in diabetic hosts, protected diabetic but not nondiabetic mice from GNB infection. Thus, rather than immunosuppression, diabetes drives lethal hyperinflammation in response to GNB by signaling through RAGE. As such, interventions to improve the outcomes from GNB infections should seek to suppress the immune response in diabetic hosts.IMPORTANCE Physicians and scientists have subscribed to the dogma that diabetes predisposes the host to worse outcomes from infections because it suppresses the immune system. This understanding was based largely on ex vivo studies of blood from patients and animals with diabetes. However, we have found that the opposite is true and worse outcomes from infection are caused by overstimulation of the immune system in response to bacteria. This overreaction occurs by simultaneous ligation of two host receptors: TLR4 and RAGE. Both signal via a common downstream messenger, MyD88, triggering hyperinflammation. In summary, contrary to hundred-year-old postulations about immune suppression in diabetic hosts, we find that diabetes instead predisposes to more severe infections because of additional inflammatory output through dual activation of MyD88 by not only TLR4 but also RAGE. It is the activation of RAGE during GNB infections in those with diabetes that accounts for their heightened susceptibility to infection compared to nondiabetic hosts.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Gram-Negative Bacterial Infections/immunology , Inflammation/immunology , Receptor for Advanced Glycation End Products/metabolism , Toll-Like Receptor 4/metabolism , Animals , Bacterial Load , Cytokines/immunology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Disease Progression , Gram-Negative Bacterial Infections/complications , Gram-Negative Bacterial Infections/metabolism , Mice , Myeloid Differentiation Factor 88/metabolism , Phagocytosis , Receptor for Advanced Glycation End Products/deficiency , Receptor for Advanced Glycation End Products/genetics , Signal Transduction , Toll-Like Receptor 4/genetics
17.
Free Radic Biol Med ; 112: 397-410, 2017 11.
Article in English | MEDLINE | ID: mdl-28826719

ABSTRACT

Oxidative stress and mitochondrial dysfunction are recognized as major contributors of cardiovascular damage in diabetes and high fat diet (HFD) fed mice. Blockade of receptor for advanced glycation end products (RAGE) attenuates vascular oxidative stress and development of atherosclerosis. We tested whether HFD-induced myocardial dysfunction would be reversed in RAGE deficiency mice, in association with changes in oxidative stress damage, mitochondrial respiration, mitochondrial fission and autophagy-lysosomal pathway. Cardiac antioxidant capacity was upregulated in RAGE-/- mice under normal diet as evidenced by increased superoxide dismutase and sirtuin mRNA expressions. Mitochondrial fragmentation and mitochondrial fission protein Drp1 and Fis1 expressions were increased in RAGE-/- mice. Autophagy-related protein expressions and cathepsin-L activity were increased in RAGE-/- mice suggesting sustained autophagy-lysosomal flux. HFD induced mitochondrial respiration defects, cardiac contractile dysfunction, disrupted mitochondrial dynamics and autophagy inhibition, which were partially prevented in RAGE-/- mice. Our results suggest that cardioprotection against HFD in RAGE-/- mice include reactivation of autophagy, as inhibition of autophagic flux by chloroquine fully abrogated beneficial myocardial effects and its stimulation by rapamycin improved myocardial function in HFD wild type mice. As mitochondrial fission is necessary to mitophagy, increased fragmentation of mitochondrial network in HFD RAGE-/- mice may have facilitated removal of damaged mitochondria leading to better mitochondrial quality control. In conclusion, modulation of RAGE pathway may improve mitochondrial damage and myocardial dysfunction in HFD mice. Attenuation of cardiac oxidative stress and maintenance of healthy mitochondria population ensuring adequate energy supply may be involved in myocardial protection against HFD.


Subject(s)
Autophagy/genetics , Cardiomyopathies/genetics , Diabetes Mellitus, Experimental/genetics , Lysosomes/metabolism , Mitochondrial Dynamics/genetics , Receptor for Advanced Glycation End Products/genetics , Animals , Autophagy/drug effects , Cardiomyopathies/chemically induced , Cardiomyopathies/drug therapy , Cardiomyopathies/pathology , Cathepsin L/genetics , Cathepsin L/metabolism , Chloroquine/pharmacology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diet, High-Fat , Dynamins/genetics , Dynamins/metabolism , Gene Expression Regulation , Lysosomes/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products/deficiency , Sirolimus/pharmacology , Sirtuins/genetics , Sirtuins/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
18.
Mol Metab ; 6(8): 897-908, 2017 08.
Article in English | MEDLINE | ID: mdl-28752053

ABSTRACT

OBJECTIVE: The hypothalamus of hypercaloric diet-induced obese animals is featured by a significant increase of microglial reactivity and its associated cytokine production. However, the role of dietary components, in particular fat and carbohydrate, with respect to the hypothalamic inflammatory response and the consequent impact on hypothalamic control of energy homeostasis is yet not clear. METHODS: We dissected the different effects of high-carbohydrate high-fat (HCHF) diets and low-carbohydrate high-fat (LCHF) diets on hypothalamic inflammatory responses in neurons and non-neuronal cells and tested the hypothesis that HCHF diets induce hypothalamic inflammation via advanced glycation end-products (AGEs) using mice lacking advanced glycation end-products (AGEs) receptor (RAGE) and/or the activated leukocyte cell-adhesion molecule (ALCAM). RESULTS: We found that consumption of HCHF diets, but not of LCHF diets, increases microgliosis as well as the presence of N(ε)-(Carboxymethyl)-Lysine (CML), a major AGE, in POMC and NPY neurons of the arcuate nucleus. Neuron-secreted CML binds to both RAGE and ALCAM, which are expressed on endothelial cells, microglia, and pericytes. On a HCHF diet, mice lacking the RAGE and ALCAM genes displayed less microglial reactivity and less neovasculature formation in the hypothalamic ARC, and this was associated with significant improvements of metabolic disorders induced by the HCHF diet. CONCLUSIONS: Combined overconsumption of fat and sugar, but not the overconsumption of fat per se, leads to excessive CML production in hypothalamic neurons, which, in turn, stimulates hypothalamic inflammatory responses such as microgliosis and eventually leads to neuronal dysfunction in the control of energy metabolism.


Subject(s)
Dietary Fats/metabolism , Dietary Sugars/metabolism , Gliosis/metabolism , Hypothalamus/metabolism , Activated-Leukocyte Cell Adhesion Molecule/genetics , Animals , Dietary Fats/adverse effects , Dietary Sugars/adverse effects , Gliosis/etiology , Glycation End Products, Advanced/metabolism , Hypothalamus/pathology , Inflammation/etiology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Rats , Receptor for Advanced Glycation End Products/deficiency , Receptor for Advanced Glycation End Products/genetics
19.
Arterioscler Thromb Vasc Biol ; 37(8): 1536-1547, 2017 08.
Article in English | MEDLINE | ID: mdl-28642238

ABSTRACT

OBJECTIVE: Diabetic subjects are at higher risk of ischemic peripheral vascular disease. We tested the hypothesis that advanced glycation end products (AGEs) and their receptor (RAGE) block angiogenesis and blood flow recovery after hindlimb ischemia induced by femoral artery ligation through modulation of immune/inflammatory mechanisms. APPROACH AND RESULTS: Wild-type mice rendered diabetic with streptozotocin and subjected to unilateral femoral artery ligation displayed increased accumulation and expression of AGEs and RAGE in ischemic muscle. In diabetic wild-type mice, femoral artery ligation attenuated angiogenesis and impaired blood flow recovery, in parallel with reduced macrophage content in ischemic muscle and suppression of early inflammatory gene expression, including Ccl2 (chemokine [C-C motif] ligand-2) and Egr1 (early growth response gene-1) versus nondiabetic mice. Deletion of Ager (gene encoding RAGE) or transgenic expression of Glo1 (reduces AGEs) restored adaptive inflammation, angiogenesis, and blood flow recovery in diabetic mice. In diabetes mellitus, deletion of Ager increased circulating Ly6Chi monocytes and augmented macrophage infiltration into ischemic muscle tissue after femoral artery ligation. In vitro, macrophages grown in high glucose display inflammation that is skewed to expression of tissue damage versus tissue repair gene expression. Further, macrophages grown in high versus low glucose demonstrate blunted macrophage-endothelial cell interactions. In both settings, these adverse effects of high glucose were reversed by Ager deletion in macrophages. CONCLUSIONS: These findings indicate that RAGE attenuates adaptive inflammation in hindlimb ischemia; underscore microenvironment-specific functions for RAGE in inflammation in tissue repair versus damage; and illustrate that AGE/RAGE antagonism may fill a critical gap in diabetic peripheral vascular disease.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Angiopathies/metabolism , Gene Deletion , Inflammation/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Peripheral Arterial Disease/metabolism , Receptor for Advanced Glycation End Products/deficiency , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Antigens, Ly/metabolism , Blood Flow Velocity , Blood Glucose/metabolism , Cell Communication , Cells, Cultured , Cellular Microenvironment , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/physiopathology , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation , Genetic Predisposition to Disease , Glycation End Products, Advanced/metabolism , Inflammation/genetics , Inflammation/physiopathology , Ischemia/genetics , Ischemia/physiopathology , Macrophages/metabolism , Mice, Knockout , Mice, Transgenic , Monocytes/metabolism , Muscle, Skeletal/metabolism , Peripheral Arterial Disease/genetics , Peripheral Arterial Disease/physiopathology , Phenotype , Receptor for Advanced Glycation End Products/genetics , Recovery of Function , Regional Blood Flow , Signal Transduction , Streptozocin , Time Factors
20.
Arterioscler Thromb Vasc Biol ; 37(4): 613-621, 2017 04.
Article in English | MEDLINE | ID: mdl-28183700

ABSTRACT

The receptor for advanced glycation endproducts (RAGE) interacts with a unique repertoire of ligands that form and collect in the tissues and circulation in diabetes mellitus, aging, inflammation, renal failure, and obesity. RAGE is expressed on multiple cell types linked to tissue perturbation in these settings. This brief review focuses on the role of RAGE in monocytes/macrophages and how RAGE ligand engagement on these cells mediates seminal changes in monocyte/macrophage migration, oxidative stress, cholesterol efflux, and pro- versus anti-inflammatory cues that signal to tissue damage. Studies using mice devoid of Ager (gene encoding RAGE) or pharmacological antagonists of RAGE are protective in animal models of diabetes mellitus, atherosclerosis, and high-fat diet-induced obesity, in least in part through key roles in monocytes/macrophages. RAGE signal transduction requires the interaction of RAGE cytoplasmic domain with the formin, DIAPH1 (diaphanous 1) and novel antagonists of this interaction show significant promise in attenuation of the maladaptive effects of RAGE ligands in cellular and in vivo models. Finally, this brief review discusses evidence for RAGE axis perturbation in human monocytes/macrophages and how tracing RAGE activity in these cells may identify target engagement biomarkers of RAGE antagonism for future clinical trials.


Subject(s)
Atherosclerosis/metabolism , Inflammation/metabolism , Macrophages/metabolism , Receptor for Advanced Glycation End Products/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Atherosclerosis/drug therapy , Atherosclerosis/etiology , Atherosclerosis/genetics , Disease Models, Animal , Humans , Inflammation/drug therapy , Inflammation/etiology , Inflammation/genetics , Ligands , Macrophages/drug effects , Mice, Knockout , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Receptor for Advanced Glycation End Products/deficiency , Receptor for Advanced Glycation End Products/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...