Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Prostate ; 84(9): 797-806, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38558412

ABSTRACT

BACKGROUND: Prostate cancer (PCa) is a common malignancy in males and obesity may play a role in its development and progression. Associations between visceral obesity measured by a body shape index (ABSI) and PCa mortality have not been thoroughly investigated. This study assessed the associations between ABSI, body mass index (BMI), and long-term PCa-specific mortality using a nationally representative US database. METHODS: This population-based longitudinal study collected data of males aged ≥40 years diagnosed with PCa and who underwent surgery and/or radiation from the National Health and Nutrition Examination Survey database 2001-2010. All included participants were followed through the end of 2019 using the National Center for Health Statistics Linked Mortality File. Associations between PCa-specific mortality, BMI, and ABSI were determined using Cox proportional hazards regression and receiver operating characteristic (ROC) curve analysis. RESULTS: Data of 294 men (representing 1,393,857 US nationals) were analyzed. After adjusting for confounders, no significant associations were found between BMI (adjusted hazard ratio [aHR] = 1.06, 95% confidence interval [CI]: 0.97-1.16, p = 0.222), continuous ABSI (aHR = 1.29, 95% CI: 0.83-2.02, p = 0.253), or ABSI in category (Q4 vs. Q1-Q3: aHR = 1.52, 95% CI: 0.72-3.24, p = 0.265), and greater risk of PCa-specific mortality. However, among participants who had been diagnosed within 4 years, the highest ABSI quartile but not in BMI was significantly associated with greater risk for PCa-specific mortality (Q4 vs. Q1-Q3: aHR = 5.34, 95% CI: 2.26-12.62, p = 0.001). In ROC analysis for this subgroup, the area under the curve of ABSI alone for predicting PCa-specific mortality was 0.638 (95% CI: 0.448-0.828), reaching 0.729 (95% CI: 0.490-0.968 when combined with other covariates. CONCLUSIONS: In US males with PCa diagnosed within 4 years, high ABSI but not BMI is independently associated with increased PCa-specific mortality.


Subject(s)
Body Mass Index , Nutrition Surveys , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Middle Aged , United States/epidemiology , Aged , Longitudinal Studies , Adult , Databases, Factual , Obesity, Abdominal/mortality , Obesity, Abdominal/epidemiology , Risk Factors
2.
Mol Nutr Food Res ; 67(9): e2200336, 2023 05.
Article in English | MEDLINE | ID: mdl-36825504

ABSTRACT

SCOPE: This study investigates the effect of epigallocatechin gallate (EGCG) on white and beige preadipocyte growth and explores the involvement of the miR-let-7a/HMGA2 pathway. METHODS AND RESULTS: 3T3-L1 and D12 cells are treated with EGCG. The effect of EGCG on cell proliferation and viability is evaluated, as well as microRNA (miRNA)-related signaling pathways. EGCG inhibits 3T3-L1 and D12 preadipocyte growth, upregulates miR-let-7a expression, and downregulates high-mobility group AT-hook 2 (HMGA2) mRNA and protein levels in a time- and dose-dependent manner. In addition, overexpression of miR-let-7a significantly inhibits the growth of 3T3-L1 and D12 cells and decreases HMGA2 mRNA and protein levels. MiR-let-7a inhibitor antagonizes the inhibitory effects of EGCG on the number and viability of 3T3-L1 and D12 cells. Furthermore, miR-let-7a inhibitor reverses the EGCG-induced increase in miR-let-7a expression levels and decrease in HMGA2 mRNA and protein levels. HMGA2 overexpression induces an increase in cell number and viability and antagonizes EGCG-suppressed cell growth and HMGA2 expression in 3T3-L1 and D12 preadipocytes. CONCLUSION: EGCG inhibits the growth of 3T3-L1 and D12 preadipocytes by modulating the miR-let-7a and HMGA2 pathways.


Subject(s)
Catechin , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Tea , Signal Transduction , Cell Proliferation , Catechin/pharmacology , RNA, Messenger
3.
Tzu Chi Med J ; 34(4): 359-366, 2022.
Article in English | MEDLINE | ID: mdl-36578640

ABSTRACT

Prostate cancer (PC) and breast cancer (BC) are the most common cancers in men and women, respectively, in developed countries. The increased incidence of PC and BC largely reflects an increase in the prevalence of obesity and metabolic syndrome. In pathological conditions involving the development and progression of PC and BC, adipose tissue plays an important role via paracrine and endocrine signaling. The increase in the amount of local adipose tissue, specifically periprostatic adipose tissue, may be a key contributor to the PC pathobiology. Similarly, breast adipose tissue secretion affects various aspects of BC by influencing tumor progression, angiogenesis, metastasis, and microenvironment. In this context, the role of white adipose tissue (WAT) has been extensively studied. However, the influence of browning of the WAT on the development and progression of PC and BC is unclear and has received less attention. In this review, we highlight that adipose tissue plays a vital role in the regulation of the tumor microenvironment in PC or BC and highlight the probable underlying mechanisms linking adipose tissue with PC or BC. We further discuss whether the browning of WAT could be a therapeutic strategy for the treatment of PC and BC.

4.
Exp Biol Med (Maywood) ; 247(18): 1670-1679, 2022 09.
Article in English | MEDLINE | ID: mdl-35894140

ABSTRACT

Green tea epigallocatechin gallate (EGCG) and microRNA (miRNA) molecules modulate obesity. Nevertheless, it is still unknown whether EGCG modulates fat cell growth via miRNA-related signaling. In this study, white preadipocytes were used to examine whether the antimitogenic effect of EGCG on fat cells is regulated by the miR-143/MAPK7 pathway. We showed that EGCG upregulated the levels of miR-143, but not miR-155, in 3T3-L1 preadipocytes. Moreover, EGCG downregulated MAPK7 mRNA and protein levels time- and dose-dependently. MAPK7 expression increased during 3T3-L1 cell proliferation. miR-143 overexpression in the absence of EGCG mimicked the effects of EGCG to suppress preadipocyte growth and MAPK7 expression, whereas knockdown of miR-143 antagonized the EGCG-altered levels of miR-143, MAPK7, and pERK1/2 and reversed the EGCG-inhibited cell growth. These findings suggest that EGCG inhibits 3T3-L1 cell growth via miR-143/MAPK7 pathway.


Subject(s)
Catechin , MicroRNAs , Mice , Animals , 3T3-L1 Cells , Tea , Catechin/pharmacology , MicroRNAs/genetics , RNA, Messenger
5.
Tzu Chi Med J ; 34(2): 113-118, 2022.
Article in English | MEDLINE | ID: mdl-35465278

ABSTRACT

Intracellular metabolites can cause critical changes in biological functions. Itaconate is perhaps the most fascinating substance in macrophages. Lipopolysaccharide can activate aconitate decarboxylase 1 and induces the generation of itaconate from the tricarboxylic acid cycle by decarboxylation of cis-aconitate. It has been reported that itaconate has beneficial effects on inflammation and oxidation. The mechanisms involved in these effects include the suppression of succinate dehydrogenase, the activation of nuclear factor E2-related factor 2 by alkylation of Kelch-like ECH-associated protein 1, suppression of aerobic glycolysis through regulation of glyceraldehyde-3-phosphate dehydrogenase and fructose-bisphosphate aldolase A, and suppression of IκBζ translation through activating transcription factor 3 activation. All of these findings elucidated the possible therapeutic implications of itaconate in inflammation-related diseases. In this review, we highlight that itaconate is a crucial molecule of the immunomodulatory response in macrophages and can regulate between immune response and cardiovascular metabolism. Furthermore, these discoveries suggest that itaconate is a very novel therapeutic molecule for the treatment of inflammation-related heart diseases.

6.
Tzu Chi Med J ; 34(2): 125-133, 2022.
Article in English | MEDLINE | ID: mdl-35465286

ABSTRACT

Sepsis is a health issue that affects millions of people worldwide. It was assumed that erythrocytes were affected by sepsis. However, in recent years, a number of studies have shown that erythrocytes affect sepsis as well. When a pathogen invades the human body, it infects the blood and organs, causing infection and sepsis-related symptoms. Pathogens change the internal environment, increasing the levels of reactive oxygen species, influencing erythrocyte morphology, and causing erythrocyte death, i.e., eryptosis. Characteristics of eryptosis include cell shrinkage, membrane blebbing, and surface exposure of phosphatidylserine (PS). Eryptotic erythrocytes increase immune cell proliferation, and through PS, attract macrophages that remove the infected erythrocytes. Erythrocyte-degraded hemoglobin derivatives and heme deteriorate infection; however, they could also be metabolized to a series of derivatives. The result that erythrocytes play an anti-infection role during sepsis provides new perspectives for treatment. This review focuses on erythrocytes during pathogenic infection and sepsis.

7.
Biomed Pharmacother ; 145: 112440, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34839254

ABSTRACT

In this study, we investigated whether the activating transcription factor 3 (ATF3) inducer ST32db, a synthetic compound with a chemical structure similar to that of native Danshen compounds, exerts an anti-obesity effect in 3T3-L1 white preadipocytes, D16 beige cells, and mice with obesity induced by a high-fat diet (HFD). The results showed that ST32db inhibited 3T3-L1 preadipocyte differentiation by inhibiting adipogenesis/lipogenesis-related gene (and protein levels) and enhancing lipolysis-related gene (and protein levels) via the activation of ß3-adrenoceptor (ß3-AR)/PKA/p38, AMPK, and ERK pathways. Furthermore, ST32db inhibited triacylglycerol accumulation in D16 adipocytes by suppressing adipogenesis/lipogenesis-related gene (and protein levels) and upregulating browning gene expression by suppressing the ß3-AR/PKA/p38, and AMPK pathways. Intraperitoneally injected ST32db (1 mg kg-1 twice weekly) inhibited body weight gain and reduced the weight of inguinal white adipose tissue (iWAT), epididymal WAT (eWAT), and mesenteric WAT, with no effects on food intake by the obese mice. The adipocyte diameter and area of iWAT and eWAT were decreased in obese mice injected with ST32db compared with those administered only HFD. In addition, ST32db significantly suppressed adipogenesis and activated lipolysis, browning, mitochondrial oxidative phosphorylation, and ß-oxidation-related pathways by suppressing the p38 pathway in the iWAT of the obese mice. These results indicated that the ATF3 inducer ST32db has therapeutic potential for reducing obesity.


Subject(s)
Activating Transcription Factor 3 , Anti-Obesity Agents , Obesity , Animals , Male , Mice , 3T3-L1 Cells , Activating Transcription Factor 3/drug effects , Activating Transcription Factor 3/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Adipose Tissue, White/drug effects , Anti-Obesity Agents/pharmacology , Cell Differentiation/drug effects , Diet, High-Fat/adverse effects , Lipolysis/drug effects , Mice, Inbred C57BL , Obesity/drug therapy , Weight Gain/drug effects
8.
Front Endocrinol (Lausanne) ; 12: 661828, 2021.
Article in English | MEDLINE | ID: mdl-34093437

ABSTRACT

Endothelins induce many biological responses, and they are composed of three peptides: ET-1, ET-2, and ET-3. Reports have indicated that ET-1 regulates cell proliferation, adipogenesis, and other cell responses and that ET-3 stimulates the growth of gastrointestinal epithelial cells and melanocytes. However, the signalling pathways of ET3 that mediate the growth of fat cells are still unclear. Using 3T3-L1 white preadipocytes, we found that ET-3 induced increases in both cell number and BrdU incorporation. Pretreatment with an ETAR antagonist (but not an ETBR antagonist) blocked the ET-3-induced increases in both cell number and BrdU incorporation. Additionally, BQ610 suppressed the ET-3-induced increases in phosphorylation of AMPK, c-JUN, and STAT3 proteins, and pretreatment with specific inhibitors of AMPK, JNK/c-JUN, or JAK/STAT3 prevented the ET-3-induced increases in phosphorylation of AMPK, c-JUN, and STAT3, respectively. Neither p38 MAPK inhibitor nor PKC inhibitor altered the effects of ET-3 on cell growth. These data suggest that ET-3 stimulates preadipocyte growth through the ETAR, AMPK, JNK/c-JUN, and STAT3 pathways. Moreover, ET-3 did not alter HIB1B brown preadipocyte and D12 beige preadipocyte growth, suggesting a preadipocyte type-dependent effect. The results of this study may help explain how endothelin mediates fat cell activity and fat cell-associated diseases.


Subject(s)
Adipocytes/cytology , Endothelin-3/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Cell Proliferation , Endothelin-3/antagonists & inhibitors , Mice , Mitogen-Activated Protein Kinases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism , Sphingomyelin Phosphodiesterase/metabolism
9.
Article in English | MEDLINE | ID: mdl-32922364

ABSTRACT

Activating transcription factor 3 (ATF3) is a stress-induced transcription factor that plays vital roles in modulating metabolism, immunity, and oncogenesis. ATF3 acts as a hub of the cellular adaptive-response network. Multiple extracellular signals, such as endoplasmic reticulum (ER) stress, cytokines, chemokines, and LPS, are connected to ATF3 induction. The function of ATF3 as a regulator of metabolism and immunity has recently sparked intense attention. In this review, we describe how ATF3 can act as both a transcriptional activator and a repressor. We then focus on the role of ATF3 and ATF3-regulated signals in modulating metabolism, immunity, and oncogenesis. The roles of ATF3 in glucose metabolism and adipose tissue regulation are also explored. Next, we summarize how ATF3 regulates immunity and maintains normal host defense. In addition, we elaborate on the roles of ATF3 as a regulator of prostate, breast, colon, lung, and liver cancers. Further understanding of how ATF3 regulates signaling pathways involved in glucose metabolism, adipocyte metabolism, immuno-responsiveness, and oncogenesis in various cancers, including prostate, breast, colon, lung, and liver cancers, is then provided. Finally, we demonstrate that ATF3 acts as a master regulator of metabolic homeostasis and, therefore, may be an appealing target for the treatment of metabolic dyshomeostasis, immune disorders, and various cancers.


Subject(s)
Activating Transcription Factor 3/genetics , Adipocytes/metabolism , Energy Metabolism/physiology , Homeostasis/physiology , Neoplasms/genetics , Activating Transcription Factor 3/metabolism , Adipocytes/pathology , Animals , Endoplasmic Reticulum Stress/physiology , Humans , Neoplasms/metabolism , Neoplasms/pathology
10.
Commun Biol ; 2: 389, 2019.
Article in English | MEDLINE | ID: mdl-31667363

ABSTRACT

Billions of people have obesity-related metabolic syndromes such as diabetes and hyperlipidemia. Promoting the browning of white adipose tissue has been suggested as a potential strategy, but a drug still needs to be identified. Here, genetic deletion of activating transcription factor 3 (ATF3-/- ) in mice under a high-fat diet (HFD) resulted in obesity and insulin resistance, which was abrogated by virus-mediated ATF3 restoration. ST32da, a synthetic ATF3 inducer isolated from Salvia miltiorrhiza, promoted ATF3 expression to downregulate adipokine genes and induce adipocyte browning by suppressing the carbohydrate-responsive element-binding protein-stearoyl-CoA desaturase-1 axis. Furthermore, ST32da increased white adipose tissue browning and reduced lipogenesis in HFD-induced obese mice. The anti-obesity efficacy of oral ST32da administration was similar to that of the clinical drug orlistat. Our study identified the ATF3 inducer ST32da as a promising therapeutic drug for treating diet-induced obesity and related metabolic disorders.


Subject(s)
Activating Transcription Factor 3/metabolism , Adipocytes, Brown/metabolism , Obesity/metabolism , 3T3-L1 Cells , Activating Transcription Factor 3/deficiency , Activating Transcription Factor 3/genetics , Adipocytes, Brown/pathology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Anti-Obesity Agents/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Body Temperature Regulation/physiology , Diet, High-Fat/adverse effects , Disease Models, Animal , Humans , Insulin Resistance , Lipogenesis/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/prevention & control , Orlistat/pharmacology , Plant Extracts/pharmacology , Plants, Medicinal/chemistry , Salvia miltiorrhiza/chemistry
11.
Int J Mol Sci ; 20(2)2019 Jan 13.
Article in English | MEDLINE | ID: mdl-30642126

ABSTRACT

Industrialized society-caused dysregular human behaviors and activities such as overworking, excessive dietary intake, and sleep deprivation lead to perturbations in the metabolism and the development of metabolic syndrome. Non-alcoholic fatty liver disease (NAFLD), the most common chronic liver disease worldwide, affects around 30% and 25% of people in Western and Asian countries, respectively, which leads to numerous medical costs annually. Insulin resistance is the major hallmark of NAFLD and is crucial in the pathogenesis and for the progression from NAFLD to non-alcoholic steatohepatitis (NASH). Excessive dietary intake of saturated fats and carbohydrate-enriched foods contributes to both insulin resistance and NAFLD. Once NAFLD is established, insulin resistance can promote the progression to the more severe state of liver endangerment like NASH. Here, we review current and potential studies for understanding the complexity between insulin-regulated glycolytic and lipogenic homeostasis and the underlying causes of NAFLD. We discuss how disruption of the insulin signal is associated with various metabolic disorders of glucoses and lipids that constitute both the metabolic syndrome and NAFLD.


Subject(s)
Glucose/metabolism , Insulin Resistance , Lipid Metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Disease Progression , Glycolysis , Homeostasis , Humans , Lipogenesis , Non-alcoholic Fatty Liver Disease/epidemiology
12.
Int J Mol Sci ; 19(11)2018 Nov 02.
Article in English | MEDLINE | ID: mdl-30400212

ABSTRACT

Traditionally, peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), a 91 kDa transcription factor, regulates lipid metabolism and long-chain fatty acid oxidation by upregulating the expression of several genes of the tricarboxylic acid cycle and the mitochondrial fatty acid oxidation pathway. In addition, PGC-1α regulates the expression of mitochondrial genes to control mitochondria DNA replication and cellular oxidative metabolism. Recently, new insights showed that several myokines such as irisin and myostatin are epigenetically regulated by PGC-1α in skeletal muscles, thereby modulating systemic energy balance, with marked expansion of mitochondrial volume density and oxidative capacity in healthy or diseased myocardia. In addition, in our studies evaluating whether PGC-1α overexpression in epicardial adipose tissue can act as a paracrine organ to improve or repair cardiac function, we found that overexpression of hepatic PGC-1α increased hepatic fatty acid oxidation and decreased triacylglycerol storage and secretion in vivo and in vitro. In this review, we discuss recent studies showing that PGC-1α may regulate mitochondrial fusion⁻fission homeostasis and affect the renal function in acute or chronic kidney injury. Furthermore, PGC-1α is an emerging protein with a biphasic role in cancer, acting both as a tumor suppressor and a tumor promoter and thus representing a new and unresolved topic for cancer biology studies. In summary, this review paper demonstrates that PGC-1α plays a central role in coordinating the gene expression of key components of mitochondrial biogenesis and as a critical metabolic regulator in many vital organs, including white and brown adipose tissue, skeletal muscle, heart, liver, and kidney.


Subject(s)
Lipid Metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Energy Metabolism , Homeostasis , Humans , Neoplasms/metabolism , Organelle Biogenesis
13.
Gen Comp Endocrinol ; 199: 46-55, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24486085

ABSTRACT

This study investigated the pathways involved in EGCG modulation of insulin-like growth factor (IGF)-stimulated glucose uptake in 3T3-L1 adipocytes. EGCG inhibited IGF-I and IGF-II stimulation of adipocyte glucose uptake with dose and time dependencies. EGCG at 20µM for 2h decreased IGF-I- and IGF-II-stimulated glucose uptake by 59% and 64%, respectively. Pretreatment of adipocytes with antibody against the EGCG receptor (also known as the 67-kDa laminin receptor; 67LR), prevented the effects of EGCG on IGF-increased glucose uptake, but pretreatment with normal rabbit immunoglobulin did not. This suggests that the 67LR mediates the anti-IGF effect of EGCG on adipocyte glucose uptake. Further analysis indicated EGCG, IGF-I, and IGF-II did not alter total levels of GLUT1 or GLUT4 protein. However, EGCG prevented the IGF-increased GLUT4 levels in the plasma membrane and blocked the IGF-decreased GLUT4 levels in low-density microsomes. Neither EGCG nor its combination with IGF altered GLUT1 protein levels in the plasma membrane and low-density microsomes. EGCG also suppressed the IGF-stimulated phosphorylation of IGF signaling molecules, PKCζ/λ, but not AKT and ERK1/2, proteins. This study suggests that EGCG suppresses IGF stimulation of 3T3-L1 adipocyte glucose uptake through inhibition of the GLUT4 translocation, but not through alterations of the GLUT1 pathway.


Subject(s)
Adipocytes/metabolism , Catechin/analogs & derivatives , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 4/metabolism , Glucose/metabolism , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor I/metabolism , Tea/chemistry , 3T3-L1 Cells , Acetylcysteine/pharmacology , Adipocytes/drug effects , Animals , Antibodies/metabolism , Catechin/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , Mice , Phosphorylation/drug effects , Protein Transport , Receptors, Laminin/metabolism , Signal Transduction/drug effects
14.
Gen Comp Endocrinol ; 196: 41-51, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24291305

ABSTRACT

Resistin is known as an adipocyte-specific hormone that can cause insulin resistance and decrease adipocyte differentiation. It can be regulated by transcriptional factors, but the possible role of forkhead transcription factor FOXO1 in regulating resistin gene expression is still unknown. Using 3T3 fibroblast and C3H10T1/2 and 3T3-L1 adipocytes, we found that transient overexpression of a non-phosphorylatable, constitutively active FOXO1, but not the wild type of FOXO1 or a DNA binding-deficient FOXO1, activated resistin promoter-directed luciferase expression. However, transient overexpression of a dominant-negative FOXO1 inactivated resistin promoter activity and reduced resistin mRNA expression. These observations indicate that the action of FOXO1 on resistin gene expression requires the activation of FOXO1 and that the effect of FOXO1 depends on the phosphorylation and dephosphorylation of FOXO1. The FOXO1 protein target sites on the resistin promoter were localized to the proximal -3545 to -787bp of 5'-flanking region of the resistin promoter. A chromatin immunoprecipitation assay also showed that FOXO1 bound the resistin promoter at nucleotide regions of -1539 to -1366bp and -1016 to -835bp, but not at the regions of -795 to -632bp. Results of this study suggest that FOXO1 transcription factor likely activates the expression of adipocyte resistin gene via direct association with the upstream resistin promoter.


Subject(s)
Adipocytes/metabolism , Forkhead Transcription Factors/physiology , Gene Expression Regulation/physiology , Promoter Regions, Genetic/genetics , Resistin/genetics , 3T3-L1 Cells , Animals , Blotting, Western , Cell Differentiation , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Forkhead Box Protein O1 , Luciferases/metabolism , Mice , Mice, Inbred C3H , NIH 3T3 Cells , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Resistin/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
Toxicol Lett ; 217(1): 42-9, 2013 Feb 13.
Article in English | MEDLINE | ID: mdl-23237828

ABSTRACT

Suppressor of cytokine signaling (SOCS)-3 can act as a regulator of energy metabolism and cytokine signaling in fat cells. It is regulated by hormones and toxicological factors. However, the action of cycloheximide on expression of adipocyte SOCS-3 gene is unknown. Using 3T3-L1 adipocytes, we found that cycloheximide up-regulated SOCS-3 mRNA expression in dose- and time-dependent manners. Treatment with actinomycin D prevented cycloheximide-stimulated SOCS-3 mRNA expression, suggesting that the effect of cycloheximide requires new mRNA synthesis. While cycloheximide was shown to increase activities of MEK1 and JNK, signaling was demonstrated to be inhibited by pretreatment with either MEK1 inhibitors U0126 and PD98059, or with the JNK inhibitor SP600125. U0126 and PD98059, respectively, reduced cycloheximide-stimulated SOCS-3 mRNA expression, but SP600125 did not antagonize cycloheximide effect. Moreover, cycloheximide was observed to up-regulate expression of other SOCS family members, such as SOCS-1, -2, -4, -5, -6, -7, and cytokine-inducible SH2-containing protein (CIS)-1 mRNAs. Such effects varied with the dosage and duration of cycloheximide treatment. These results imply the functional MEK1/ERK-mediated pathway is necessary for the cycloheximide stimulation of SOCS-3 gene expression.


Subject(s)
Adipocytes, White/drug effects , Cycloheximide/pharmacology , Down-Regulation/drug effects , MAP Kinase Signaling System/drug effects , Protein Synthesis Inhibitors/pharmacology , Suppressor of Cytokine Signaling Proteins/metabolism , Up-Regulation/drug effects , 3T3-L1 Cells , Adipocytes, White/metabolism , Animals , Cell Line , Dactinomycin/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Osmolar Concentration , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Rats , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Time Factors
16.
Mol Nutr Food Res ; 56(4): 580-92, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22495985

ABSTRACT

SCOPE: This study investigated the pathways involved in epigallocatechin gallate (EGCG) modulation of insulin-like growth factor (IGF)-I-stimulated and IGF-II-stimulated mitogenesis in 3T3-L1 preadipocytes. METHODS AND RESULTS: We found that this process was dose and time dependent, and caused by suppression of IGF-I-stimulated and IGF-II-stimulated phosphorylation of p66Shc and mitogen-activated protein kinase (MAPK) pathway proteins, including MEK1 kinase (RAF1), extracellular signal-regulated protein kinase (ERK) kinase (MEK1), and ERK 1 and ERK 2 (ERK1/2), but not phospho-Jun-N-terminal kinase, protein kinase B, p52Shc, or p46Shc. Furthermore, EGCG inhibited the IGF-I-stimulated phosphorylation of the IGF-I receptor-beta (IGF-IR ß), the association of IGF-IR with the p66Shc protein, and the IGF-II-stimulated associations of the IGF-II receptor with G(αi-2) and p66Shc proteins, suggesting that EGCG selectively affects particular types of Shc and MAPK family members. Pretreatment with antiserum against the EGCG receptor (also known as the 67-kDa laminin receptor; 67LR), but not with an adenosine monophosphate (AMP)-activated protein kinase (AMPK) inhibitor, prevented the inhibitory actions of EGCG on IGF-I- and IGF-II-stimulated ERK1/2 phosphorylation and subsequent preadipocyte proliferation. CONCLUSION: The results of this study suggest that EGCG mediates anti-IGF-I and anti-IGF-II signals in preadipocyte mitogenesis via the 67LR but not the AMPK pathway.


Subject(s)
Adipocytes/drug effects , Catechin/analogs & derivatives , Cell Proliferation/drug effects , Insulin-Like Growth Factor II/antagonists & inhibitors , Insulin-Like Growth Factor I/antagonists & inhibitors , Receptors, Laminin/metabolism , 3T3-L1 Cells , Adenosine Monophosphate/metabolism , Animals , Antioxidants/pharmacology , Catechin/pharmacology , Immunoprecipitation , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Plant Extracts/pharmacology , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 2/metabolism , Tea/chemistry
17.
Planta Med ; 76(15): 1694-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20455202

ABSTRACT

Insulin and (-)-epigallocatechin gallate (EGCG) are reported to regulate obesity and fat accumulation, respectively. This study investigated the pathways involved in EGCG modulation of insulin-stimulated glucose uptake in 3T3-L1 and C3H10T1/2 adipocytes. EGCG inhibited insulin stimulation of adipocyte glucose uptake in a dose- and time-dependent manner. The concentration of EGCG that decreased insulin-stimulated glucose uptake by 50-60% was approximately 5-10 µM for a period of 2 h. At 10 µM, EGCG and gallic acid were more effective than (-)-epicatechin, (-)-epigallocatechin, and (-)-epicatechin 3-gallate. We identified the EGCG receptor [also known as the 67-kDa laminin receptor (67LR)] in fat cells and extended the findings for this study to clarify whether EGCG-induced changes in insulin-stimulated glucose uptake in adipocytes could be mediated through the 67LR. Pretreatment of adipocytes with a 67LR antibody, but not normal rabbit immunoglobulin, prevented the effects of EGCG on insulin-increased glucose uptake. This suggests that the 67LR mediates the effect of EGCG on insulin-stimulated glucose uptake in adipocytes. Moreover, pretreatment with an AMP-activated protein kinase (AMPK) inhibitor, such as compound C, but not with a glutathione (GSH) activator, such as N-acetyl-L-cysteine (NAC), blocked the antiinsulin effect of EGCG on adipocyte glucose uptake. These data suggest that EGCG exerts its anti-insulin action on adipocyte glucose uptake via the AMPK, but not the GSH, pathway. The results of this study possibly support that EGCG mediates fat content.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Catechin/analogs & derivatives , Glucose/metabolism , Insulin/pharmacology , Receptors, Laminin/physiology , Tea/chemistry , Animals , Catechin/chemistry , Catechin/isolation & purification , Catechin/pharmacology , Mice , Mice, Inbred C3H , NIH 3T3 Cells
18.
Am J Physiol Cell Physiol ; 297(1): C121-32, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19176763

ABSTRACT

Insulin and (-)-epigallocatechin gallate (EGCG) have been reported to regulate fat cell mitogenesis and adipogenesis, respectively. This study investigated the pathways involved in EGCG modulation of insulin-stimulated mitogenesis in 3T3-L1 preadipocytes. EGCG inhibited insulin stimulation of preadipocyte proliferation in a dose- and time-dependent manner. EGCG also suppressed insulin-stimulated phosphorylation of the insulin receptor-beta, insulin receptor (IR) substrates 1 and 2 (IRS1 and IRS2), and mitogen-activated protein kinase pathway proteins, RAF1, MEK1/2, and ERK1/2, but not JNK. Furthermore, EGCG inhibited the association of IR with the IRS1 and IRS2 proteins, but not with the IRS4 protein. These data suggest that EGCG selectively affects particular types of IRS and MAPK family members. Generally, EGCG was more effective than epicatechin, epicatechin gallate, and epigallocatechin in modulating insulin-stimulated mitogenic signaling. We identified the EGCG receptor [also known as the 67-kDa laminin receptor (67LR)] in fat cells and found that its expression was sensitive to growth phase, tissue type, and differentiation state. Pretreatment of preadipocytes with 67LR antiserum prevented the effects of EGCG on insulin-stimulated phosphorylation of IRS2, RAF1, and ERK1/2 and insulin-stimulated preadipocyte proliferation (cell number and bromodeoxyuridine incorporation). Moreover, EGCG tended to increase insulin-stimulated associations between the 67LR and IR, IRS1, IRS2, and IRS4 proteins. These data suggest that EGCG mediates anti-insulin signaling in preadipocyte mitogenesis via the 67LR pathway.


Subject(s)
Adipocytes/drug effects , Anti-Obesity Agents/pharmacology , Camellia sinensis , Catechin/analogs & derivatives , Cell Proliferation/drug effects , Insulin/metabolism , Receptors, Laminin/drug effects , Signal Transduction/drug effects , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Anti-Obesity Agents/isolation & purification , Camellia sinensis/chemistry , Catechin/isolation & purification , Catechin/pharmacology , Dose-Response Relationship, Drug , Immune Sera , Insulin Receptor Substrate Proteins/metabolism , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Proto-Oncogene Proteins c-raf/metabolism , Receptors, Laminin/immunology , Receptors, Laminin/metabolism , Time Factors
19.
Mol Nutr Food Res ; 53(3): 349-60, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19065584

ABSTRACT

Green tea (-)-epigallocatechin-3-gallate (EGCG) is known as to regulate obesity and fat cell activity. However, little information is known about the effects of EGCG on oxidative reactive oxygen species (ROS) of fat cells. Using 3T3-L1 preadipocytes and adipocytes, we found that EGCG increased ROS production in dose- and time-dependent manners. The concentration of EGCG that increased ROS levels by 180-500% was approximately 50 muM for a range of 8-16 h of treatment. In contrast, EGCG dose- and time-dependently decreased the amount of intracellular glutathione (GSH) levels. EGCG was more effective than (-)-epicatechin, (-)-epicatechin-3-gallate, and (-)-epigallocatechin in changing ROS and GSH levels. This suggests a catechin-specific effect. To further examine the relation of GSH to ROS as altered by EGCG, we observed that exposure of preadipocytes and adipocytes to N-acetyl-L-cysteine (a GSH precursor) blocked the EGCG-induced increases in ROS levels and decreases in GSH levels. These observations suggest a GSH-dependent effect of EGCG on ROS production. While EGCG was demonstrated to alter levels of ROS and GSH, its signaling was altered by an EGCG receptor (the so-called 67 kDa laminin receptor(67LR)) antiserum, but not by normal rabbit serum. These data suggest that EGCG mediates GSH and ROS levels via the 67LR pathway.


Subject(s)
Adipocytes/chemistry , Catechin/analogs & derivatives , Glutathione/analysis , Reactive Oxygen Species/analysis , Receptors, Laminin/physiology , Tea/chemistry , 3T3-L1 Cells , Acetylcysteine/pharmacology , Adipocytes/drug effects , Animals , Antibodies/pharmacology , Catechin/pharmacology , Glutathione/antagonists & inhibitors , Mice , Receptors, Laminin/immunology
20.
Am J Physiol Cell Physiol ; 294(6): C1542-51, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18417718

ABSTRACT

Resistin is known as an adipocyte-specific secretory hormone that can cause insulin resistance and decrease adipocyte differentiation. It can be regulated by sexual hormones. Whether environmental estrogens regulate the production of resistin is still not clear. Using 3T3-L1 adipocytes, we found that octylphenol upregulated resistin mRNA expression in dose- and time-dependent manners. The concentration of octylphenol that increased resistin mRNA levels by 50% was approximately 100 nM within 6 h of treatment. The basal half-life of resistin mRNA induced by actinomycin D was lengthened by octylphenol treatment, suggesting that octylphenol decreases the rate of resistin mRNA degradation. In addition, octylphenol stimulated resistin protein expression and release. The basal half-life of resistin protein induced by cycloheximide was lengthened by octylphenol treatment, suggesting that octylphenol decreases the rate of resistin protein degradation. While octylphenol was shown to increase activities of the estrogen receptor (ER) and MEK1, signaling was demonstrated to be blocked by pretreatment with either ICI-182780 (an ERalpha antagonist) or U-0126 (a MEK1 inhibitor), in which both inhibitors prevented octylphenol-stimulated phosphorylation of ERK. These results imply that ERalpha and ERK are necessary for the octylphenol stimulation of resistin mRNA expression. Moreover, U-0126 antagonized the octylphenol-increased resistin protein expression and release. These data suggest that the way octylphenol signaling increases resistin protein levels is similar to that by which it increases resistin mRNA levels; it is likely mediated through an ERK-dependent pathway. In vivo, octylphenol increased adipose resistin mRNA expression and serum resistin and glucose levels, supporting its in vitro effect.


Subject(s)
Adipocytes/drug effects , Endocrine Disruptors/pharmacology , Estrogen Receptor alpha/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Phenols/pharmacology , Resistin/metabolism , Signal Transduction/drug effects , 3T3-L1 Cells , Adipocytes/enzymology , Adipocytes/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Animals , Benzhydryl Compounds , Blood Glucose/drug effects , Butadienes/pharmacology , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Dose-Response Relationship, Drug , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/metabolism , Fulvestrant , Half-Life , Leptin/genetics , Leptin/metabolism , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , Mice , Nitriles/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Synthesis Inhibitors/pharmacology , RNA Stability , RNA, Messenger/metabolism , Resistin/blood , Resistin/genetics , Time Factors , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...