Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters











Publication year range
1.
Inflammopharmacology ; 29(4): 1061-1074, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34185201

ABSTRACT

The liver acts as a manufacturing unit for the production of fetuin-A, which is essential for various physiological characteristics. Scientific research has shown that a moderate upward push in fetuin-A serum levels is associated with a confirmed non-alcoholic fatty liver disease (NAFLD) diagnosis. Fetuin-A modulation is associated with a number of pathophysiological variables that cause liver problems, including insulin receptor signaling deficiencies, adipocyte dysfunction, hepatic inflammation, fibrosis, triacylglycerol production, macrophage invasion, and TLR4 activation. The focus of the present review is on the various molecular pathways, and genetic relevance of mRNA expression of fetuin-A which is correlated with progression of NAFLD. The other major area of exploration in the present review is based on the new targets for the modulation of fetuin-A, like calorie restriction and novel pharmacological agents, such as rosuvastatin, metformin, and pioglitazone which are successfully implicated in the management of various liver-related complications.


Subject(s)
Liver/metabolism , Liver/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , alpha-2-HS-Glycoprotein/biosynthesis , Adiponectin/antagonists & inhibitors , Adiponectin/biosynthesis , Adiponectin/genetics , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Caloric Restriction/methods , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Insulin Resistance/physiology , Liver/drug effects , Metformin/pharmacology , Metformin/therapeutic use , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/genetics , alpha-2-HS-Glycoprotein/antagonists & inhibitors , alpha-2-HS-Glycoprotein/genetics
2.
Int J Mol Sci ; 21(17)2020 Sep 03.
Article in English | MEDLINE | ID: mdl-32899357

ABSTRACT

Animal and human mechanistic studies have consistently shown an association between obesity and Alzheimer's disease (AD). AD, a degenerative brain disease, is the most common cause of dementia and is characterized by the presence of extracellular amyloid beta (Aß) plaques and intracellular neurofibrillary tangles disposition. Some studies have recently demonstrated that Aß and tau cannot fully explain the pathophysiological development of AD and that metabolic disease factors, such as insulin, adiponectin, and antioxidants, are important for the sporadic onset of nongenetic AD. Obesity prevention and treatment can be an efficacious and safe approach to AD prevention. Adiponectin is a benign adipokine that sensitizes the insulin receptor signaling pathway and suppresses inflammation. It has been shown to be inversely correlated with adipose tissue dysfunction and may enhance the risk of AD because a range of neuroprotection adiponectin mechanisms is related to AD pathology alleviation. In this study, we summarize the recent progress that addresses the beneficial effects and potential mechanisms of adiponectin in AD. Furthermore, we review recent studies on the diverse medications of adiponectin that could possibly be related to AD treatment, with a focus on their association with adiponectin. A better understanding of the neuroprotection roles of adiponectin will help clarify the precise underlying mechanism of AD development and progression.


Subject(s)
Adiponectin/antagonists & inhibitors , Adiponectin/metabolism , Alzheimer Disease/drug therapy , Anti-Obesity Agents/therapeutic use , Gene Expression Regulation , Neuroprotective Agents/therapeutic use , Obesity/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Humans , Obesity/metabolism , Obesity/pathology
3.
Sci Rep ; 9(1): 14189, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31578352

ABSTRACT

Interactions between multiple myeloma (MM) and bone marrow (BM) are well documented to support tumour growth, yet the cellular mechanisms underlying pain in MM are poorly understood. We have used in vivo murine models of MM to show significant induction of nerve growth factor (NGF) by the tumour-bearing bone microenvironment, alongside other known pain-related characteristics such as spinal glial cell activation and reduced locomotion. NGF was not expressed by MM cells, yet bone stromal cells such as osteoblasts expressed and upregulated NGF when cultured with MM cells, or MM-related factors such as TNF-α. Adiponectin is a known MM-suppressive BM-derived factor, and we show that TNF-α-mediated NGF induction is suppressed by adiponectin-directed therapeutics such as AdipoRON and L-4F, as well as NF-κB signalling inhibitor BMS-345541. Our study reveals a further mechanism by which cellular interactions within the tumour-bone microenvironment contribute to disease, by promoting pain-related properties, and suggests a novel direction for analgesic development.


Subject(s)
Adiponectin/genetics , Multiple Myeloma/drug therapy , Nerve Growth Factor/genetics , Pain/drug therapy , Tumor Necrosis Factor-alpha/genetics , Adiponectin/antagonists & inhibitors , Animals , Bone Marrow/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imidazoles/pharmacology , Mice , Multiple Myeloma/complications , Multiple Myeloma/genetics , Multiple Myeloma/pathology , NF-kappa B/antagonists & inhibitors , Neuroglia/metabolism , Neuroglia/pathology , Osteoblasts/drug effects , Pain/complications , Pain/genetics , Pain/pathology , Peptides/pharmacology , Piperidines/pharmacology , Quinoxalines/pharmacology , Stromal Cells/drug effects , Tumor Microenvironment/drug effects
4.
Int J Mol Sci ; 20(4)2019 Feb 20.
Article in English | MEDLINE | ID: mdl-30791536

ABSTRACT

Pulmonary hypertension is an umbrella term including many different disorders causing an increase of the mean pulmonary arterial pressure (mPAP) ≥ 25 mmHg. Recent data revealed a strong association between obesity and pulmonary hypertension. Adiponectin is a protein synthetized by the adipose tissue with pleiotropic effects on inflammation and cell proliferation, with a potential protective role on the pulmonary vasculature. Both in vivo and in vitro studies documented that adiponectin is an endogenous modulator of NO production and interferes with AMP-activated protein kinase (AMPK) activation, mammalian target of rapamycin (mTOR), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κß) signaling preventing endothelial dysfunction and proliferation. Furthermore, adiponectin ameliorates insulin resistance by mediating the biological effects of peroxisome proliferator-activated receptor-gamma (PPARγ). Therefore, adiponectin modulation emerged as a theoretical target for the treatment of pulmonary hypertension, currently under investigation. Recently, consistent data showed that hypoglycemic agents targeting PPARγ as well as renin⁻angiotensin system inhibitors and mineralocorticoid receptor blockers may influence pulmonary hemodynamics in different models of pulmonary hypertension.


Subject(s)
Hypertension, Pulmonary/etiology , Obesity/complications , Adiponectin/antagonists & inhibitors , Adiponectin/genetics , Adiponectin/metabolism , Animals , Biomarkers , Disease Susceptibility , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Molecular Targeted Therapy , Myocytes, Smooth Muscle/metabolism , Obesity/genetics , Obesity/metabolism , Signal Transduction
5.
Arthritis Res Ther ; 20(1): 245, 2018 Oct 30.
Article in English | MEDLINE | ID: mdl-30376894

ABSTRACT

BACKGROUND: Different adiponectin isoforms appear to be differentially involved in the pathogenesis of various diseases. The purpose of this study was to generate monoclonal antibodies (mAbs) specific to different adiponectin isoforms and investigate whether these mAbs have potential as therapeutic agents for such diseases. METHODS: Hybridoma cells producing monoclonal antibodies were generated and screened using enzyme-linked immunosorbent assay and Western blotting for the production of mAbs recognizing human adiponectin isoforms. RESULTS: The mAb from hybridoma clone KH7-41 recognized both the middle molecular weight (MMW) (hexamer) and low molecular weight (LMW) (trimer) isoforms of adiponectin in human serum, whereas the KH7-33 mAb detected only MMW (hexamer) adiponectin. The KH4-8 clone recognized both the high molecular weight (HMW) (multimer) and MMW adiponectin isoforms. However, in mouse and rat sera, the abovementioned antibodies recognized only the MMW isomer. These mAbs also recognized adiponectin in various human tissues, such as lung, kidney, and adipose tissues, although the three mAbs had different staining intensities. The mAb from clone KH4-8 effectively inhibited increases in interleukin-6 (IL-6) and IL-8 expression in recombinant adiponectin-stimulated human osteoblasts and human umbilical vein endothelial cells. Also, the mAbs KH7-33 and KH4-8 significantly ameliorated rheumatic symptoms in a collagen-induced arthritis mouse model. This result suggests that these mAb treatments may ameliorate adiponectin-mediated inflammatory response. CONCLUSIONS: mAbs against human adiponectin isomers can potentially be developed as therapeutic antibodies to target specific detrimental isoforms of adiponectin while maintaining the functions of beneficial isoforms.


Subject(s)
Adiponectin/blood , Antibodies, Monoclonal/administration & dosage , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/drug therapy , Drug Delivery Systems/methods , Adiponectin/antagonists & inhibitors , Aged , Animals , Female , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Middle Aged , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/blood
6.
Cell Mol Biol (Noisy-le-grand) ; 64(4): 71-77, 2018 Mar 31.
Article in English | MEDLINE | ID: mdl-29631686

ABSTRACT

Obesity is global problem that contributes to disease, and is partly caused by fast-food, high-fat diets. Much attention has been focused on developing anti-obesity foods and chemical materials from natural sources. Seaweed has bioactive properties that influence immune activity and have anti-cancer and anti-obesity effects. Laminaria japonica is a widely consumed seaweed, and has been promoted as a health food in Korea. The bioactive properties of L. japonica include anti-cancer, anti-diabetic, and anti-inflammation effects. Most Laminaria japonica are distributed in a simple processing form such as drying, and their availability is very low. Therefore, various types of functional products can be developed if they can be applied to foods through functionalization using fermentation techniques. It is a structural problem that is the most problematic in seaweed processing. In this study, we used fermented Laminaria japonica. To increase physiological activity, fermentation treatment was performed to loosen the structure, thereby increasing the activity of the glycoprotein. First, we screened the anti-obesity potential of an L. japonica fermentation extract (LJF) using 3T3-L1 adipocyte cells. We determined cytotoxicity using an MTS assay and measured LJF for its ability to affect adipogenesis through glucose uptake, triglyceride levels, and Oil Red O staining. We confirmed that LJF inhibited adipocyte differentiation. CCAAT/enhancer-binding proteins α/ß (C/EBP-α/ß) and peroxisome proliferator-activated receptor-γ (PPAR-γ) are involved in the early and late stages of adipocyte differentiation. LJF significantly reduced the expression levels of C/EBP-α/ß and PPAR-γ and decreased the concentration of adiponectin. Thus, our results suggest that LJF inhibits adipogenesis in 3T3-L1 cells, and may be valuable for its anti-obesity effects.


Subject(s)
Adipocytes/drug effects , Anti-Obesity Agents/pharmacology , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Proteins/genetics , Complex Mixtures/pharmacology , Laminaria/chemistry , PPAR gamma/genetics , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/drug effects , Adipogenesis/genetics , Adiponectin/antagonists & inhibitors , Adiponectin/genetics , Adiponectin/metabolism , Animals , Anti-Obesity Agents/chemistry , CCAAT-Enhancer-Binding Protein-beta/antagonists & inhibitors , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/drug effects , Complex Mixtures/chemistry , Fermentation , Gene Expression Regulation , Lipid Metabolism/drug effects , Mice , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Signal Transduction
7.
Cell Physiol Biochem ; 43(1): 172-181, 2017.
Article in English | MEDLINE | ID: mdl-28848139

ABSTRACT

BACKGROUND/AIMS: Adiponectin (Apn) is a multifunctional adipokine that circulates as several oligomeric complexes in the blood stream. Previous reports showed that several conserved lysine residues within the N-terminal collagenous domain of Apn are modified by hydroxylation and glycosylation. Here, we investigated the potential roles of post-translational modifications of Apn on the function of human vascular smooth muscle cells (VSMCs). METHODS: Blood samples of 92 coronary artery disease (CAD) patients and 20 healthy volunteers were collected and total and high molecular weight (HMW) Apn concentration and glycosylation were analyzed. RESULTS: The results revealed that total and HMW Apn derived from blood samples of CAD patients with severe stenosis significantly increased, however the glycosylation of HMW Apn significantly decreased. Functional studies of human VSMCs revealed that glycosylated Apn significantly inhibited the oxidized LDL-induced lipid accumulation, proliferation and migration of VSMCs, whereas non-glycosylated Apn had no inhibitory effects. CONCLUSION: Taken together, these data suggest that glycosylation of Apn is critically involved in regulating function against atherosclerosis by inhibiting lipid accumulation and proliferation and migration of VSMCs.


Subject(s)
Adiponectin/metabolism , Coronary Artery Disease/pathology , Adiponectin/antagonists & inhibitors , Adiponectin/blood , Adiponectin/genetics , Case-Control Studies , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cholesterol/analysis , Coronary Artery Disease/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique, Indirect , Glycosylation , Humans , Lipoproteins, LDL/pharmacology , Male , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Severity of Illness Index
8.
Biochem Biophys Res Commun ; 490(2): 202-208, 2017 08 19.
Article in English | MEDLINE | ID: mdl-28606474

ABSTRACT

Adiponectin is a protein secreted by white adipocytes that plays an important role in insulin action, energy homeostasis and the development of atherosclerosis. The intracellular localization and trafficking of GLUT4 and leptin in adipocytes has been well studied, but little is known regarding the intracellular trafficking of adiponectin. Recent studies have demonstrated that constitutive adiponectin secretion is dependent on PIP2 levels and the integrity of cortical F-actin. Non-muscle myosin II is an actin-based motor that is associated with membrane vesicles and participates in vesicular trafficking in mammalian cells. Therefore, we investigated the role of myosin II in the trafficking and secretion of adiponectin in 3T3-L1 adipocytes. Confocal microscopy revealed that myosin IIA and IIB were dispersed throughout the cytoplasm of the adipocyte. Both myosin isoforms were localized in the Golgi/TGN region as evidenced by colocalization with the cis-Golgi marker, p115 and the trans-Golgi marker, γ-adaptin. Inhibition of myosin II activity by blebbistatin or actin depolymerization by latrunculin B dispersed myosin IIA and IIB towards the periphery while significantly inhibiting adiponectin secretion. Therefore, the constitutive trafficking and secretion of adiponectin in 3T3-L1 adipocytes occurs by an actin-dependent mechanism that involves the actin-based motors, myosin IIA and IIB.


Subject(s)
Adiponectin/metabolism , Nonmuscle Myosin Type IIA/metabolism , Nonmuscle Myosin Type IIB/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adiponectin/antagonists & inhibitors , Animals , Biological Transport/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Heterocyclic Compounds, 4 or More Rings/pharmacology , Mice , Nonmuscle Myosin Type IIA/antagonists & inhibitors , Nonmuscle Myosin Type IIB/antagonists & inhibitors , Structure-Activity Relationship , Thiazolidines/pharmacology
9.
J Mol Endocrinol ; 58(4): 155-165, 2017 05.
Article in English | MEDLINE | ID: mdl-28219936

ABSTRACT

It has been reported that ischemic preconditioning (IPC) and adiponectin (APN) are cardioprotective in many cardiovascular disorders. However, whether APN mediates the effect of IPC on myocardial injury has not been elucidated. This study was conducted to investigate whether IPC affects myocardial ischemic injury by increasing APN expression. Male adult rats with cardiac knockdowns of APN and its receptors via intramyocardial small-interfering RNA injection were subjected to IPC and then myocardial infarction (MI) at 24 h after IPC. Globular APN (gAd) was injected at 10 min before MI. APN mRNA and protein levels in myocardium as well as the plasma APN concentration were markedly high at 6 and 12 h after IPC. IPC ameliorated myocardial injury as evidenced by improved cardiac functions and a reduced infarct size. Compared with the control MI group, rats in the IPC + MI group had elevated levels of left ventricular ejection fraction and fractional shortening and a smaller MI size (P < 0.05). However, the aforementioned protective effects were ameliorated in the absence of APN and APN receptors, followed by the inhibition of AMP-activated protein kinase (AMPK) phosphorylation, but reversed by gAd treatment in wild-type rats, and AMPK phosphorylation increased (P < 0.05). Overall, our results suggest that the cardioprotective effects of IPC are partially due to upregulation of APN and provide a further insight into IPC-mediated signaling effects.


Subject(s)
Adiponectin/genetics , Ischemic Preconditioning, Myocardial , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Receptors, Adiponectin/genetics , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adiponectin/antagonists & inhibitors , Adiponectin/metabolism , Animals , Gene Expression Regulation , Heart Function Tests , Male , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Adiponectin/antagonists & inhibitors , Receptors, Adiponectin/metabolism , Signal Transduction , Stroke Volume/physiology
10.
Eur J Nutr ; 56(2): 879-891, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26700221

ABSTRACT

PURPOSE: DNA methylation is one of the most extensively studied mechanisms within epigenetics, and it is suggested that diet-induced changes in methylation status could be involved in energy metabolism regulation. Conjugated linoleic acid (CLA) and calcium supplementation counteract body weight gain, particularly under a high-fat (HF) diet, in adult mice. The aim was to determine whether the modulation of DNA methylation pattern in target genes and tissues could be an underlying mechanism of action. METHODS: Mice (C57BL/6J) were divided into five groups according to diet and treatment: normal fat as the control group (12 % kJ content as fat), HF group (43 % kJ content as fat), HF + CLA (6 mg CLA/day), HF + calcium (12 g/kg of calcium) and HF with both compounds. Gene expression and methylation degree of CpG sites in promoter sequences of genes involved in fatty acid metabolism, including adiponectin (Adipoq), stearoyl-CoA desaturase (Scd1) and fatty acid synthase (Fasn), were determined by bisulphite sequencing in liver and epididymal white adipose tissue. RESULTS: Results showed that the methylation profile of promoters was significantly altered by dietary supplementation in a gene- and tissue-specific manner, whereas only slight changes were observed in the HF group. Furthermore, changes in specific CpG sites were also associated with an overall healthier metabolic profile, in particular for calcium-receiving groups. CONCLUSIONS: Both CLA and calcium were able to modify the methylation pattern of genes involved in energy balance in adulthood, which opens a novel area for increasing efficiency in body weight management strategies.


Subject(s)
Anti-Obesity Agents/therapeutic use , Calcium, Dietary/therapeutic use , DNA Methylation , Dietary Supplements , Epigenesis, Genetic , Linoleic Acids, Conjugated/therapeutic use , Obesity/prevention & control , Adiponectin/antagonists & inhibitors , Adiponectin/genetics , Adiponectin/metabolism , Adipose Tissue, White/enzymology , Adipose Tissue, White/metabolism , Animals , CpG Islands , Diet, High-Fat/adverse effects , Energy Metabolism , Fatty Acid Synthase, Type I/antagonists & inhibitors , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Liver/enzymology , Liver/metabolism , Male , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Organ Specificity , Promoter Regions, Genetic , RNA, Messenger/metabolism , Stearoyl-CoA Desaturase/chemistry , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Weight Gain
11.
J Endocrinol Invest ; 40(4): 437-445, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27914036

ABSTRACT

PURPOSE: Prolactin regulatory element-binding protein (PREB), a member of the WD-repeat protein family, has been recognized as a transcriptional factor that regulates prolactin promoter activity in the anterior pituitary of rats. PREB is expressed not only in the pituitary but also in various other tissues, including the adipose tissue. Previous studies have shown that PREB acts as a transcriptional regulator and suppresses the expression of the adiponectin gene in cultured 3T3L1 preadipocytes. The aim of this study was to further examine the potential role of PREB in adipose tissue in vivo. METHODS: Transgenic mice that overexpressing PREB (PREB transgenic mice) were generated. Insulin resistance was evaluated in PREB transgenic mice using glucose and insulin tolerance tests. Adiponectin expression in the adipose tissue was examined by western blot analysis and quantitative polymerase chain reaction (qPCR). The expression levels of stearoyl-CoA desaturase (Scd) and adiponectin receptor 2(ADIPOR2) were quantified by qPCR. RESULTS: Glucose and insulin tolerance tests revealed insulin resistance in PREB transgenic mice. Serum adiponectin and leptin concentrations were decreased. Adiponectin gene expression was decreased in the adipose tissue, which was confirmed by the downregulation of the adiponectin-dependent hepatic Scd gene and upregulation of the ADIPOR2 gene in the liver of PREB transgenic mice. We also found that pioglitazone, an agonist for the peroxisome proliferator-activated receptor-r, improved the insulin resistance in the PREB transgenic mice after a 10-day feeding period. CONCLUSIONS: These results demonstrated that PREB might contribute to the regulation of adiponectin gene expression in vivo.


Subject(s)
Adiponectin/antagonists & inhibitors , DNA-Binding Proteins/physiology , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/physiology , Insulin Resistance , Transcription Factors/physiology , Adiponectin/genetics , Adiponectin/metabolism , Animals , Humans , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
12.
Ann N Y Acad Sci ; 1380(1): 183-194, 2016 09.
Article in English | MEDLINE | ID: mdl-27441783

ABSTRACT

Gastroesophageal reflux disease (GERD) has become the most commonly seen gastrointestinal disorder in outpatient clinics. In the United States, around 20% of the general population experience heartburn on a weekly basis. Although clinical complaints can be mild or moderate, patients with GERD may develop further complications, such as peptic strictures, Barrett's esophagus (BE), and even esophageal adenocarcinoma. Pathologically, GERD is developed as a result of chronic and enhanced exposure of the esophageal epithelium to noxious gastric refluxate. In this review article, we provide an overview of GERD and then focus on the roles of stromal cells, interleukin 4, and adiponectin in GERD and BE. The importance of inflammation and immunomodulators in GERD pathogenesis is highlighted. Targeting the immunomodulators or inflammation in general may improve the therapeutic outcome of GERD, in particular, in those refractory to proton pump inhibitors.


Subject(s)
Adiponectin/metabolism , Gastroesophageal Reflux/complications , Gastroesophageal Reflux/immunology , Immunologic Factors/metabolism , Interleukin-4/metabolism , Adiponectin/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Gastroesophageal Reflux/diagnosis , Humans , Immunologic Factors/antagonists & inhibitors , Interleukin-4/antagonists & inhibitors , Stromal Cells/drug effects , Stromal Cells/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism
13.
Life Sci ; 132: 49-54, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25896661

ABSTRACT

AIMS: Adipocytes play a critical role in energy balance. Growth of fat tissue is achieved via an increase in adipocyte mass and the formation of newly differentiated adipocytes from precursor cells. Understanding the cellular and molecular mechanisms of adipocyte differentiation is crucial for the study of obesity- and fat-related diseases. The present study was designed to study whether small heterodimer partner-interacting leucine zipper protein (SMILE), a novel co-repressor, could regulate differentiation of adipocyte in 3T3-L1 cells. MATERIALS AND METHODS: Treatment of endoplasmic stress inducers, thapsigargin and tunicamycin, inhibited adipocyte differentiation, stimulated Smile mRNA expression, and repressed the expression of adiponectin (Adipoq) in 3T3-L1 pre-adipocyte. Overexpression of SMILE in 3T3-L1 cells decreased the expression of the mRNA encoding Adipoq, a major marker of adipocytes, significantly. Furthermore, knockdown of SMILE recovered the thapsigargin-mediated repression of Adipoq transcription. Co-immunoprecipitation experiments revealed that SMILE interacted physically with PPARγ in 3T3-L1 cells. In addition, chromatin immunoprecipitation experiments revealed that SMILE suppressed the binding affinity of PPARγ for the Adipoq promoter. KEY FINDINGS: We demonstrate that SMILE controls adipocyte differentiation by regulating the transactivity of peroxisome proliferator-activated receptor γ (PPARγ). SIGNIFICANCE: These findings demonstrate that SMILE represses adipocyte differentiation by regulating PPARγ transactivity; hence, SMILE is a potential regulator of PPARγ-related diseases.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , Basic-Leucine Zipper Transcription Factors/pharmacology , Cell Differentiation/drug effects , PPAR gamma/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adiponectin/antagonists & inhibitors , Adiponectin/metabolism , Animals , Azo Compounds , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Blotting, Western , Chromatin Immunoprecipitation , DNA Primers/genetics , Gene Knockdown Techniques , Genetic Vectors/genetics , Mice , Real-Time Polymerase Chain Reaction
14.
Circulation ; 131(16): 1392-404, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25696921

ABSTRACT

BACKGROUND: Phosphorylative desensitization of G-protein-coupled receptors contributes significantly to post-myocardial infarction (MI) remodeling and heart failure (HF). Here, we determined whether adiponectin receptors (AdipoRs) 1 and 2 (the 7-transmembrane domain-containing receptors mediating adiponectin functions) are phosphorylatively modified and functionally impaired after MI. METHODS AND RESULTS: Post-MI HF was induced by coronary artery occlusion. Receptor phosphorylation, kinase expression, and adiponectin function were determined via in vivo, ex vivo, and in vitro models. AdipoR1 and AdipoR2 are not phosphorylated in the normal heart. However, AdipoR1 was significantly phosphorylated after MI, peaking at 7 days and remaining significantly phosphorylated thereafter. The extent of post-MI AdipoR1 phosphorylation positively correlated with the expression level of GPCR kinase (GRK) 2, the predominant GRK isoform upregulated in the failing heart. Cardiac-specific GRK2 knockout virtually abolished post-MI AdipoR1 phosphorylation, whereas virus-mediated GRK2 overexpression significantly phosphorylated AdipoR1 and blocked adiponectin metabolic-regulatory/anti-inflammatory signaling. Mass spectrometry identified serine-7, threonine-24, and threonine-53 (residues located in the n-terminal intracellular AdipoR1 region) as the GRK2 phosphorylation sites. Ex vivo experiments demonstrated that adenosine monophosphate-activated protein kinase activation and the anti-tumor necrosis factor-α effect of adiponectin were significantly inhibited in cardiomyocytes isolated from nonischemic area 7 days after MI. In vivo experiments demonstrated that acute adiponectin administration-induced cardiac GLUT4 translocation and endothelial nitric oxide synthase phosphorylation were blunted 7 days after MI. Continuous adiponectin administration beginning 7 days after MI failed to protect the heart from adverse remodeling and HF progression. Finally, cardiac-specific GRK2 knockdown restored the cardioprotective effect of adiponectin. CONCLUSION: AdipoR1 is phosphorylatively modified and desensitized by GRK2 in failing cardiomyocytes, contributing to post-MI remodeling and HF progression.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/physiology , Heart Failure/enzymology , Protein Processing, Post-Translational , Receptors, Adiponectin/metabolism , AMP-Activated Protein Kinases/metabolism , Adiponectin/antagonists & inhibitors , Adiponectin/pharmacology , Animals , Cells, Cultured , G-Protein-Coupled Receptor Kinase 2/deficiency , G-Protein-Coupled Receptor Kinase 2/genetics , Genetic Therapy , Genetic Vectors/therapeutic use , Glucose Transporter Type 4/metabolism , Heart Failure/pathology , Heart Failure/therapy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/complications , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Phosphorylation , Recombinant Fusion Proteins/metabolism , Transduction, Genetic
15.
Am J Physiol Endocrinol Metab ; 306(12): E1418-30, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24780611

ABSTRACT

Adiponectin (APN), the most abundant adipocyte-secreted adipokine, regulates energy homeostasis and exerts well-characterized insulin-sensitizing properties. The peripheral or central effects of APN regulating bone metabolism are beginning to be explored but are still not clearly understood. In the present study, we found that APN-knockout (APN-KO) mice fed a normal diet exhibited decreased trabecular structure and mineralization and increased bone marrow adiposity compared with wild-type (WT) mice. APN intracerebroventricular infusions decreased uncoupling protein 1 (UCP1) expression in brown adipose tissue, epinephrine and norepinephrine serum levels, and osteoclast numbers, whereas osteoblast osteogenic marker expression and trabecular bone mass increased in APN-KO and WT mice. In addition, centrally administered APN increased hypothalamic tryptophan hydroxylase 2 (TPH2), cocaine- and amphetamine-regulated transcript (CART), and 5-hydroxytryptamine (serotonin) receptor 2C (Htr2C) expressions but decreased hypothalamic cannabinoid receptor-1 expression. Treatment of immortalized mouse neurons with APN demonstrated that APN-mediated effects on TPH2, CART, and Htr2C expression levels were abolished by downregulating adaptor protein containing pleckstrin homology domain, phosphotyrosine domain, and leucine zipper motif (APPL)-1 expression. Pharmacological increase in sympathetic activity stimulated adipogenic differentiation of bone marrow stromal cells (BMSC) and reversed APN-induced expression of the lysine-specific demethylases involved in regulating their commitment to the osteoblastic lineage. In conclusion, we found that APN regulates bone metabolism via central and peripheral mechanisms to decrease sympathetic tone, inhibit osteoclastic differentiation, and promote osteoblastic commitment of BMSC.


Subject(s)
Adiponectin/pharmacology , Bone Density Conservation Agents/pharmacology , Bone Marrow/drug effects , Bone and Bones/drug effects , Osteogenesis/drug effects , Peptide Fragments/pharmacology , Recombinant Proteins/pharmacology , Adiponectin/antagonists & inhibitors , Adiponectin/chemistry , Adiponectin/genetics , Adiposity/drug effects , Animals , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/antagonists & inhibitors , Bone Density Conservation Agents/chemistry , Bone Marrow/metabolism , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Cell Line , Gene Expression Regulation/drug effects , Gene Silencing , Humans , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/metabolism , Infusions, Intraventricular , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Conformation , Radiography , Random Allocation , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry
16.
J Mol Cell Cardiol ; 74: 183-92, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24852843

ABSTRACT

The adipocyte-secreted hormone adiponectin (APN) exerts protective effects on the heart under stress conditions. Recent studies have demonstrated that APN induces a marked Ca(2+) influx in skeletal muscle. However, whether APN modulates [Ca(2+)]i activity, especially [Ca(2+)]i transients in cardiomyocytes, is still unknown. This study was designed to determine whether APN modulates [Ca(2+)]i transients in cardiomyocytes. Adult male wild-type (WT) and APN knockout (APN KO) mice were subjected to myocardial ischemia/reperfusion (I/R, 30min/30min) injury. CaMKII-PLB phosphorylation and SR Ca(2+)-ATPase (SERCA2) activity were downregulated in I/R hearts of WT mice and further decreased in those of APN KO mice. Both the globular domain of APN and full-length APN significantly reversed the decrease in CaMKII-PLB phosphorylation and SERCA2 activity in WT and APN KO mice. Interestingly, compared with WT littermates, single myocytes isolated from APN KO mice had remarkably decreased [Ca(2+)]i transients, cell shortening, and a prolonged Ca(2+) decay rate. Further examination revealed that APN enhances SERCA2 activity via CaMKII-PLB signaling. In in vivo and in vitro experiments, both APN receptor 1/2 and S1P were necessary for the APN-stimulated CaMKII-PLB-SERCA2 activation. In addition, S1P activated CaMKII-PLB signaling in neonatal cardiomyocytes in a dose dependent manner and improved [Ca(2+)]i transients in APN KO myocytes via the S1P receptor (S1PR1/3). Further in vivo experiments revealed that pharmacological inhibition of S1PR1/3 and SERCA2 siRNA suppressed APN-mediated cardioprotection during I/R. These data demonstrate that S1P is a novel regulator of SERCA2 that activates CaMKII-PLB signaling and mediates APN-induced cardioprotection.


Subject(s)
Adiponectin/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Lysophospholipids/metabolism , Myocardial Reperfusion Injury/genetics , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum/metabolism , Sphingosine/analogs & derivatives , Adiponectin/antagonists & inhibitors , Adiponectin/metabolism , Adiponectin/pharmacology , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Gene Expression Regulation , Lysophospholipids/pharmacology , Male , Mice , Mice, Knockout , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phosphorylation , Protein Structure, Tertiary , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Adiponectin/genetics , Receptors, Adiponectin/metabolism , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction , Sphingosine/metabolism , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors
17.
Endocrinology ; 155(5): 1806-16, 2014 May.
Article in English | MEDLINE | ID: mdl-24564394

ABSTRACT

Adiponectin, an adipocyte-derived hormone, regulates glucose and lipid metabolism. It is also antiinflammatory. During obesity, adiponectin levels and sensitivity are reduced. Whereas the action of adiponectin in the periphery is well established the neuroendocrine role of adiponectin is largely unknown. To address this we analyzed the expression of adiponectin and the 2 adiponectin receptors (AdipoR1 and AdipoR2) in response to fasting and to diet-induced and genetic obesity. We also investigated the acute impact of adiponectin on central regulation of glucose homeostasis. Adiponectin (1 µg) was injected intracerebroventricularly (ICV), and glucose tolerance tests were performed in dietary and genetic obese mice. Finally, the influence of ICV adiponectin administration on central signaling cascades regulating glucose homeostasis and on markers of hypothalamic inflammation was assessed. Gene expression of adiponectin was down-regulated whereas AdipoR1 was up-regulated in the arcuate nucleus of fasted mice. High-fat (HF) feeding increased AdipoR1 and AdipoR2 gene expression in this region. In mice on a HF diet and in leptin-deficient mice acute ICV adiponectin improved glucose tolerance 60 minutes after injection, whereas normoglycemia in control mice was unaffected. ICV adiponectin increased pAKT, decreased phospho-AMP-activated protein kinase, and did not change phospho-signal transducer and activator of transcription 3 immunoreactivity. In HF-fed mice, ICV adiponectin reversed parameters of hypothalamic inflammation and insulin resistance as determined by the number of phospho-glycogen synthase kinase 3 ß(Ser9) and phospho-c-Jun N-terminal kinase (Thr183/Tyr185) immunoreactive cells in the arcuate nucleus and ventromedial hypothalamus. This study demonstrates that the insulin-sensitizing properties of adiponectin are at least partially based on a neuroendocrine mechanism that involves centrally synthesized adiponectin.


Subject(s)
Adiponectin/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Glucose Intolerance/prevention & control , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Obesity/physiopathology , Adiponectin/administration & dosage , Adiponectin/adverse effects , Adiponectin/antagonists & inhibitors , Adiponectin/genetics , Adiponectin/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/pathology , Diet, High-Fat/adverse effects , Glucose Intolerance/etiology , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Injections, Intraventricular , Leptin/genetics , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Receptors, Adiponectin/biosynthesis , Receptors, Adiponectin/genetics , Receptors, Adiponectin/metabolism , Signal Transduction/drug effects , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/immunology , Ventromedial Hypothalamic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/pathology
18.
Eur J Nutr ; 53(3): 951-61, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24173963

ABSTRACT

PURPOSE: To evaluate the long-term consequences of TNFα inhibitors on body composition and fat distribution, as well as changes in serum adipokines in patients with rheumatoid arthritis (RA) or ankylosing spondylitis (AS). METHODS: Eight patients with RA and twelve with AS requiring a TNFα inhibitor were prospectively followed for 2 years. Body composition was evaluated by dual X-ray absorptiometry and included measurements of total fat mass, lean mass, fat in the gynoid and android regions, and visceral fat. Serum leptin, total and high molecular weight (HMW) adiponectin, resistin, and ghrelin were also assessed. RESULTS: There was a significant gain in body mass index (p = 0.05) and a tendency for weight (p = 0.07), android fat (p = 0.07), and visceral fat (p = 0.059) increase in patients with RA, while in AS, total fat mass significantly increased (p = 0.02) with a parallel weight gain (p = 0.07). When examining the whole population of patients, we observed after 2 years a significant increase in body weight (+1.9%; p = 0.003), body mass index (+2.5%; p = 0.004), total fat mass (+11.1%; p = 0.007), and fat in the android region (+18.3%; p = 0.02). There was a substantial, albeit nonsignificant gain in visceral fat (+24.3%; p = 0.088). Lean mass and gynoid fat were not modified. No major changes were observed for serum leptin, total adiponectin, and ghrelin, while HMW adiponectin and the HMW/total adiponectin ratio tended to decrease (-15.2%, p = 0.057 and -9.3%, p = 0.067, respectively). Resistin decreased significantly (-22.4%, p = 0.01). CONCLUSIONS: Long-term TNFα inhibition in RA and AS is associated with a significant gain in fat mass, with a shift to the android (visceral) region. This fat redistribution raises questions about its influence on the cardiovascular profile of patients receiving these treatments.


Subject(s)
Abdominal Fat/drug effects , Adiposity/drug effects , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Arthritis, Rheumatoid/drug therapy , Spondylitis, Ankylosing/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Abdominal Fat/immunology , Abdominal Fat/metabolism , Abdominal Fat/pathology , Adiponectin/antagonists & inhibitors , Adiponectin/blood , Adiponectin/metabolism , Adult , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Body Mass Index , Drug Monitoring , Female , Follow-Up Studies , Humans , Immunoglobulin G/adverse effects , Immunoglobulin G/therapeutic use , Insulin Resistance , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/pathology , Male , Middle Aged , Prospective Studies , Resistin/antagonists & inhibitors , Resistin/blood , Resistin/metabolism , Spondylitis, Ankylosing/immunology , Spondylitis, Ankylosing/metabolism , Spondylitis, Ankylosing/pathology , Weight Gain/drug effects
19.
Kidney Blood Press Res ; 37(6): 657-66, 2013.
Article in English | MEDLINE | ID: mdl-24356556

ABSTRACT

BACKGROUND: We hypothesized that plasma levels of brain natriuretic peptide (BNP) and N-terminal pro-BNP (NT-proBNP) would be elevated, and adiponectin concentrations reduced, in patients with atherosclerotic renal artery stenosis (ARAS) and that BNPs might be used to identify patients who would benefit from percutaneous transluminal renal angioplasty (PTRA). METHODS: Data were collected before renal angiography in 91 patients with hypertension and suspected ARAS (significant ARAS; n=47, and non-RAS; n=44) and in 20 healthy controls (C). In ARAS patients analyses were repeated four weeks after PTRA. RESULTS: Ambulatory systolic blood pressure (ASBP) was significantly elevated in the ARAS group vs. both C and non-RAS groups. Baseline plasma BNP and NT-proBNP levels were significantly elevated, and adiponectin concentrations reduced, in the ARAS group vs. C but not vs. the non-RAS group. One month after PTRA, ASBP was reduced vs. baseline (149±16 to 139±15 mm p<0.01). Brain natriuretic peptides were not significantly affected by PTRA. CONCLUSIONS: Patients with ARAS showed elevated of BNP and NT-proBNP concentrations, and reduced levels of adiponectin, compared to healthy controls but not vs. hypertensive individuals without RAS. Our data do no support the use of BNP analyses in the identification of ARAS patients who will have a beneficial blood pressure response to PTRA.


Subject(s)
Angioplasty , Atherosclerosis/blood , Atherosclerosis/therapy , Natriuretic Peptide, Brain/blood , Renal Artery Obstruction/blood , Renal Artery Obstruction/therapy , Adiponectin/antagonists & inhibitors , Adiponectin/blood , Aged , Angioplasty/methods , Atherosclerosis/diagnostic imaging , Biomarkers/blood , Female , Humans , Male , Middle Aged , Radiography , Renal Artery/diagnostic imaging , Renal Artery Obstruction/diagnostic imaging
20.
J Mammary Gland Biol Neoplasia ; 18(3-4): 321-32, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24136336

ABSTRACT

Obesity is a recognized risk factor for breast cancer development and poorer response to therapy. Two major fat tissue-derived adipokines, leptin and adiponectin have been implicated in mammary carcinogenesis. Leptin appears to promote breast cancer progression through activation of mitogenic, antiapoptotic, and metastatic pathways, while adiponectin may restrict tumorigenic processes primarily by inhibiting cell metabolism. Furthermore, adiponectin is known to counteract detrimental leptin effects in breast cancer models. Thus, therapeutic inhibition of pro-neoplastic leptin pathways and reactivation of anti-neoplastic adiponectin signaling may benefit breast cancer patients, especially the obese subpopulation. This review focuses on current experimental strategies aiming at leptin and adiponectin pathways in breast cancer models. Novel leptin receptor antagonists and adiponectin receptor agonists as well as other compounds for therapeutic modulation of adipokine pathways are discussed in detail, including potential pharmacological advantages and limitations of these approaches.


Subject(s)
Adiponectin/antagonists & inhibitors , Breast Neoplasms/drug therapy , Leptin/antagonists & inhibitors , Molecular Targeted Therapy , Adiponectin/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Leptin/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL