Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
Mol Cell Biol ; : 1-14, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38804232

ABSTRACT

Betaine-homocysteine S-methyltransferase (BHMT) is one of the most abundant proteins in the liver and regulates homocysteine metabolism. However, the molecular mechanisms underlying Bhmt transcription have not yet been elucidated. This study aimed to assess the molecular mechanisms underlying Bhmt transcription and the effect of BHMT deficiency on metabolic functions in the liver mediated by liver receptor homolog-1 (LRH-1). During fasting, both Bhmt and Lrh-1 expression increased in the liver of Lrh-1f/f mice; however, Bhmt expression was decreased in LRH-1 liver specific knockout mice. Promoter activity analysis confirmed that LRH-1 binds to a specific site in the Bhmt promoter region. LRH-1 deficiency was associated with elevated production of reactive oxygen species (ROS), lipid peroxidation, and mitochondrial stress in hepatocytes, contributing to hepatic triglyceride (TG) accumulation. In conclusion, this study suggests that the absence of an LRH-1-mediated decrease in Bhmt expression promotes TG accumulation by increasing ROS levels and inducing mitochondrial stress. Therefore, LRH-1 deficiency not only leads to excess ROS production and mitochondrial stress in hepatocytes, but also disrupts the methionine cycle. Understanding these regulatory pathways may pave the way for novel therapeutic interventions against metabolic disorders associated with hepatic lipid accumulation.

2.
Exp Mol Med ; 56(4): 1001-1012, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38622198

ABSTRACT

Sterol regulatory element-binding protein (SREBP)-1c is involved in cellular lipid homeostasis and cholesterol biosynthesis and is highly increased in nonalcoholic steatohepatitis (NASH). However, the molecular mechanism by which SREBP-1c regulates hepatic stellate cells (HSCs) activation in NASH animal models and patients have not been fully elucidated. In this study, we examined the role of SREBP-1c in NASH and the regulation of LCN2 gene expression. Wild-type and SREBP-1c knockout (1cKO) mice were fed a high-fat/high-sucrose diet, treated with carbon tetrachloride (CCl4), and subjected to lipocalin-2 (LCN2) overexpression. The role of LCN2 in NASH progression was assessed using mouse primary hepatocytes, Kupffer cells, and HSCs. LCN2 expression was examined in samples from normal patients and those with NASH. LCN2 gene expression and secretion increased in CCl4-induced liver fibrosis mice model, and SREBP-1c regulated LCN2 gene transcription. Moreover, treatment with holo-LCN2 stimulated intracellular iron accumulation and fibrosis-related gene expression in mouse primary HSCs, but these effects were not observed in 1cKO HSCs, indicating that SREBP-1c-induced LCN2 expression and secretion could stimulate HSCs activation through iron accumulation. Furthermore, LCN2 expression was strongly correlated with inflammation and fibrosis in patients with NASH. Our findings indicate that SREBP-1c regulates Lcn2 gene expression, contributing to diet-induced NASH. Reduced Lcn2 expression in 1cKO mice protects against NASH development. Therefore, the activation of Lcn2 by SREBP-1c establishes a new connection between iron and lipid metabolism, affecting inflammation and HSCs activation. These findings may lead to new therapeutic strategies for NASH.


Subject(s)
Iron , Lipocalin-2 , Liver Cirrhosis , Mice, Knockout , Non-alcoholic Fatty Liver Disease , Sterol Regulatory Element Binding Protein 1 , Animals , Humans , Male , Mice , Carbon Tetrachloride/pharmacology , Disease Models, Animal , Gene Expression Regulation , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Iron/metabolism , Lipocalin-2/metabolism , Lipocalin-2/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/genetics , Liver Cirrhosis/chemically induced , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics
3.
Pflugers Arch ; 476(2): 151-161, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37940681

ABSTRACT

Pancreatic beta cells utilize Ca2+ to secrete insulin in response to glucose. The glucose-dependent increase in cytosolic Ca2+ concentration ([Ca2+]C) activates a series of insulin secretory machinery in pancreatic beta cells. Therefore, the amount of insulin secreted in response to glucose is determined in a [Ca2+]C-dependent manner, at least within a moderate range. However, the demand for insulin secretion may surpass the capability of beta cells. Abnormal elevation of [Ca2+]C levels beyond the beta-cell endurance capacity can damage them by inducing endoplasmic reticulum (ER) stress and cell death programs such as apoptosis. Therefore, while Ca2+ is essential for the insulin secretory functions of beta cells, it could affect their survival at pathologically higher levels. Because an increase in beta-cell [Ca2+]C is inevitable under certain hazardous conditions, understanding the regulatory mechanism for [Ca2+]C is important. Therefore, this review discusses beta-cell function, survival, ER stress, and apoptosis associated with intracellular and ER Ca2+ homeostasis.


Subject(s)
Insulin-Secreting Cells , Insulin-Secreting Cells/metabolism , Calcium Signaling , Insulin/metabolism , Endoplasmic Reticulum/metabolism , Calcium/metabolism , Glucose/metabolism
4.
Life Sci ; 332: 122107, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37739164

ABSTRACT

AIMS: Prolonged high levels of cytokines, glucose, or free fatty acids are associated with diabetes, elevation of cytosolic Ca2+ concentration ([Ca2+]C), and depletion of Ca2+ concentration in the endoplasmic reticulum (ER) of pancreatic beta cells. This Ca2+ imbalance induces ER stress and apoptosis. Lupenone, a lupan-type triterpenoid, is beneficial in diabetes; however, its mechanism of action is yet to be clarified. This study evaluated the protective mechanism of lupenone against thapsigargin-induced ER stress and apoptosis in pancreatic beta cells. MATERIALS AND METHODS: MIN6, INS-1, and native mouse islet cells were used. Western blot for protein expressions, measurement of [Ca2+]C, and in vivo glucose tolerance test were mainly performed. KEY FINDINGS: Thapsigargin increased the protein levels of cleaved caspase 3, cleaved PARP, and the phosphorylated form of JNK, ATF4, and CHOP. Thapsigargin increased the interaction between stromal interaction molecule1 (Stim1) and Orai1, enhancing store-operated calcium entry (SOCE). SOCE is further activated by protein tyrosine kinase 2 (Pyk2), which is Ca2+-dependent and phosphorylates the tyrosine residue at Y361 in Stim1. Lupenone inhibited thapsigargin-mediated Pyk2 activation, suppressed [Ca2+]C, ER stress, and apoptosis. Lupenone restored impaired glucose-stimulated insulin secretion effectuated by thapsigargin and glucose intolerance in a low-dose streptozotocin-induced diabetic mouse model. SIGNIFICANCE: These results suggested that lupenone attenuated thapsigargin-induced ER stress and apoptosis by inhibiting SOCE; this may be due to the hindrance of Pyk2-mediated Stim1 tyrosine phosphorylation. In beta cells that are inevitably exposed to frequent [Ca2+]C elevation, the attenuation of abnormally high SOCE would be beneficial for their survival.


Subject(s)
Diabetes Mellitus , Insulin-Secreting Cells , Lupanes , Triterpenes , Animals , Mice , Apoptosis , Calcium/metabolism , Cell Line , Diabetes Mellitus/metabolism , Endoplasmic Reticulum Stress , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 2/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Phosphorylation , Thapsigargin/adverse effects , Triterpenes/metabolism , Tyrosine/metabolism , Lupanes/pharmacology
5.
J Yeungnam Med Sci ; 40(1): 4-11, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35368155

ABSTRACT

Caloric restriction is a popular approach to treat obesity and its associated chronic illnesses but is difficult to maintain for a long time. Intermittent fasting is an alternative and easily applicable dietary intervention for caloric restriction. Moreover, intermittent fasting has beneficial effects equivalent to those of caloric restriction in terms of body weight control, improvements in glucose homeostasis and lipid profiles, and anti-inflammatory effects. In this review, the beneficial effects of intermittent fasting are discussed.

6.
J Adv Res ; 45: 1-13, 2023 03.
Article in English | MEDLINE | ID: mdl-35659922

ABSTRACT

INTRODUCTION: Sterol regulatory element binding protein (SREBP) cleavage-associating protein (SCAP) is a sterol-regulated escort protein that translocates SREBPs from the endoplasmic reticulum to the Golgi apparatus, thereby activating lipid metabolism and cholesterol synthesis. Although SCAP regulates lipid metabolism in metabolic tissues, such as the liver and muscle, the effect of macrophage-specific SCAP deficiency in adipose tissue macrophages (ATMs) of patients with metabolic diseases is not completely understood. OBJECTIVES: Here, we examined the function of SCAP in high-fat/high-sucrose diet (HFHS)-fed mice and investigated its role in the polarization of classical activated macrophages in adipose tissue. METHODS: Macrophage-specific SCAP knockout (mKO) mice were generated through crossbreeding lysozyme 2-cre mice with SCAP floxed mice which were then fed HFHS for 12 weeks. Primary macrophages were derived from bone marrow cells and analyzed further. RESULTS: We found that fat accumulation and the appearance of proinflammatory M1 macrophages were both higher in HFHS-fed SCAP mKO mice relative to floxed control mice. We traced the effect to a defect in the lipopolysaccharide-mediated increase in SREBP-1a that occurs in control but not SCAP mKO mice. Mechanistically, SREBP-1a increased expression of cholesterol 25-hydroxylase transcription, resulting in an increase in the production of 25-hydroxycholesterol (25-HC), an endogenous agonist of liver X receptor alpha (LXRα) which increased expression of cholesterol efflux to limit cholesterol accumulation and M1 polarization. In the absence of SCAP mediated activation of SREBP-1a, increased M1 macrophage polarization resulted in reduced cholesterol efflux downstream from 25-HC-dependent LXRα activation. CONCLUSION: Overall, the activation of the SCAP-SREBP-1a pathway in macrophages may provide a novel therapeutic strategy that ameliorates obesity by controlling cholesterol homeostasis in ATMs.


Subject(s)
Insulin Resistance , Mice , Animals , Sterol Regulatory Element Binding Protein 1 , Intracellular Signaling Peptides and Proteins , Cholesterol , Obesity
7.
J Cancer ; 13(8): 2570-2583, 2022.
Article in English | MEDLINE | ID: mdl-35711835

ABSTRACT

Aims: Ribosomal protein L17 (RPL17), a 60S subunit component, is up-regulated in colorectal cancer (CRC). However, its oncogenic role in CRC progression remains unexplored. Thus, we aimed to investigate the effect of RPL17 targeting on CRC in vitro and in vivo and whether RPL17 gained an extra-ribosomal function during CRC development. Methods: RPL17-specific siRNAs complexed with cationic lipids were transfected to CRC cells to silence target gene expression and then real-time RT-PCR and western blotting were applied to observe the change of expression or activity of genes or proteins of interest. Cell proliferation assay, clonogenic assay and cell cycle analysis were used to determine the in vitro effects of RPL17siRNAs on CRC cell growth, and a subcutaneous xenograft assay was applied to test the effect of RPL17siRNAs on in vivo tumor growth. RNA sequencing and western blotting were used to investigate the underlying mechanisms. Sphere-forming assay, invasion assay and migration assay were used to evaluate the effects of RPL17siRNAs on CRC stemness. Results: siRNA-mediated inhibition of RPL17 expression suppressed CRC cell growth and long-term colony formation by inducing apoptotic cell death. Similarly, targeting RPL17 effectively suppressed tumor formation in a mouse xenograft model. RNA sequencing of RPL17-silenced CRC cells revealed the same directional regulation of 159 (93 down- and 66 up-regulated) genes. Notably, NIMA-related kinase 2 (NEK2), which functionally cooperates with extracellular-regulated protein kinase (ERK) and plays a pivotal role in mitotic progression and stemness maintenance, was down-regulated. RPL17 silencing reduced NEK2, ß-catenin, and p-ERK protein levels. These molecular alterations reflected the reduction in sphere-forming capacity, expression of stem cell marker genes, migration, and invasion. Reversely, RPL17 overexpression increased the ability of long-term colony formation, migration, and invasion. Conclusion: Our findings indicate that RPL17 promotes CRC proliferation and stemness via the ERK and NEK2/ß-catenin signaling axis, and targeting RPL17 could be the next molecular strategy for both primary CRC treatment and prevention of secondary tumor formation.

8.
BMB Rep ; 54(9): 476-481, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34488925

ABSTRACT

Liver receptor homolog-1 (LRH-1) has emerged as a regulator of hepatic glucose, bile acid, and mitochondrial metabolism. However, the functional mechanism underlying the effect of LRH-1 on lipid mobilization has not been addressed. This study investigated the regulatory function of LRH-1 in lipid metabolism in maintaining a normal liver physiological state during fasting. The Lrh-1f/f and LRH-1 liver-specific knockout (Lrh-1LKO) mice were either fed or fasted for 24 h, and the liver and serum were isolated. The livers were used for qPCR, western blot, and histological analysis. Primary hepatocytes were isolated for immunocytochemistry assessments of lipids. During fasting, the Lrh-1LKO mice showed increased accumulation of triglycerides in the liver compared to that in Lrh-1f/f mice. Interestingly, in the Lrh-1LKO liver, decreases in perilipin 5 (PLIN5) expression and genes involved in ß-oxidation were observed. In addition, the LRH-1 agonist dialauroylphosphatidylcholine also enhanced PLIN5 expression in human cultured HepG2 cells. To identify new target genes of LRH-1, these findings directed us to analyze the Plin5 promoter sequence, which revealed -1620/-1614 to be a putative binding site for LRH-1. This was confirmed by promoter activity and chromatin immunoprecipitation assays. Additionally, fasted Lrh-1f/f primary hepatocytes showed increased co-localization of PLIN5 in lipid droplets (LDs) compared to that in fasted Lrh-1LKO primary hepatocytes. Overall, these findings suggest that PLIN5 might be a novel target of LRH-1 to mobilize LDs, protect the liver from lipid overload, and manage the cellular needs during fasting. [BMB Reports 2021; 54(9): 476-481].


Subject(s)
Liver/metabolism , Perilipin-5/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Triglycerides/metabolism , Animals , Binding Sites , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Lipid Droplets/metabolism , Male , Mice , Mice, Knockout , Perilipin-5/chemistry , Perilipin-5/genetics , Promoter Regions, Genetic , Protein Binding , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics
9.
Mol Cell ; 81(18): 3820-3832.e7, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34233158

ABSTRACT

A metabolic imbalance between lipid synthesis and degradation can lead to hepatic lipid accumulation, a characteristic of patients with non-alcoholic fatty liver disease (NAFLD). Here, we report that high-fat-diet-induced sterol regulatory element-binding protein (SREBP)-1c, a key transcription factor that regulates lipid biosynthesis, impairs autophagic lipid catabolism via altered H2S signaling. SREBP-1c reduced cystathionine gamma-lyase (CSE) via miR-216a, which in turn decreased hepatic H2S levels and sulfhydration-dependent activation of Unc-51-like autophagy-activating kinase 1 (ULK1). Furthermore, Cys951Ser mutation of ULK1 decreased autolysosome formation and promoted hepatic lipid accumulation in mice, suggesting that the loss of ULK1 sulfhydration was directly associated with the pathogenesis of NAFLD. Moreover, silencing of CSE in SREBP-1c knockout mice increased liver triglycerides, confirming the connection between CSE, autophagy, and SREBP-1c. Overall, our results uncover a 2-fold mechanism for SREBP-1c-driven hepatic lipid accumulation through reciprocal activation and inhibition of hepatic lipid biosynthesis and degradation, respectively.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Fatty Liver/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Autophagy , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/physiology , Cell Line, Tumor , Diet, High-Fat/adverse effects , Fatty Liver/physiopathology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism/physiology , Lipids/physiology , Lipogenesis , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Signal Transduction/physiology , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/physiology , Triglycerides/metabolism
10.
Cancers (Basel) ; 13(5)2021 Mar 02.
Article in English | MEDLINE | ID: mdl-33801424

ABSTRACT

Big data analysis has revealed the upregulation of cell division cycle associated 8 (CDCA8) in human hepatocellular carcinoma (HCC) and its poorer survival outcome. However, the functions of CDCA8 during HCC development remain unknown. Here, we demonstrate in vitro that CDCA8 silencing inhibits HCC cell growth and long-term colony formation and migration through the accumulation of the G2/M phase cell population. Conversely, CDCA8 overexpression increases the ability to undergo long-term colony formation and migration. RNA sequencing and bioinformatic analysis revealed that CDCA8 knockdown led to the same directional regulation in 50 genes (25 down- and 25 upregulated). It was affirmed based on protein levels that CDCA8 silencing downregulates the levels of cyclin B1 and p-cdc2 and explains how it could induce G2/M arrest. The same condition increased the protein levels of tumor-suppressive ATF3 and GADD34 and inactivated AKT/ß-catenin signaling, which plays an important role in cell growth and stemness, reflecting a reduction in sphere-forming capacity. Importantly, it was demonstrated that the extent of CDCA8 expression is much greater in CD133+ cancer stem cells than in CD133- cancer cells, and that CDCA8 knockdown decreases levels of CD133, p-Akt and ß-catenin and increases levels of ATF3 and GADD34 in the CD133+ cancer stem cell (CSC) population. These molecular changes led to the inhibition of cell growth and sphere formation in the CD133+ cell population. Targeting CDCA8 also effectively suppressed tumor growth in a murine xenograft model, showing consistent molecular alterations in tumors injected with CDCA8siRNA. Taken together, these findings indicate that silencing CDCA8 suppresses HCC growth and stemness via restoring the ATF3 tumor suppressor and inactivating oncogenic AKT/ß-catenin signaling, and that targeting CDCA8 may be the next molecular strategy for both primary HCC treatment and the prevention of metastasis or recurrence.

11.
Biomedicines ; 10(1)2021 Dec 30.
Article in English | MEDLINE | ID: mdl-35052752

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a major global health issue. The development of T2DM is gradual and preceded by the pre-diabetes mellitus (pre-DM) stage, which often remains undiagnosed. This study aimed to identify novel pre-DM biomarkers in a high-fat diet (HFD)-induced pre-DM mouse model. Male C57BL/6J mice were fed either a chow diet or HFD for 12 weeks. Serum and liver samples were isolated in a time-dependent manner. Semi-quantitative assessment of secretory cytokines was performed by cytokine array analysis, and 13 cytokines were selected for further analysis based on the changes in expression levels in the pre-DM and T2DM stages. HFD-fed mice gained body weight and exhibited high serum lipid, liver enzyme, glucose, and insulin levels during the progression of pre-DM to T2DM. The mRNA expression of inflammatory and lipogenic genes was elevated in HFD-fed mice The mRNA expression of Fc receptor, IgG, low affinity Iib, lectin, galactose binding, soluble 1, vascular cell adhesion molecule 1, insulin-like growth factor binding protein 5, and growth arrest specific 6 was elevated in the pre-DM, which was confirmed by measuring protein levels. Our study identified novel pre-DM biomarkers that may help to delay or prevent the progression of T2DM.

12.
Redox Biol ; 37: 101749, 2020 10.
Article in English | MEDLINE | ID: mdl-33080438

ABSTRACT

Obesity is regarded as an abnormal expansion and excessive accumulation of fat mass in white adipose tissue. The involvement of oxidative stress in the development of obesity is still unclear. Although mainly present in peroxisomes, catalase scavenges intracellular H2O2 at toxic levels. Therefore, we used catalase-knockout (CKO) mice to elucidate the involvement of excessive H2O2 in the development of obesity. CKO mice with C57BL/6J background gained more weight with higher body fat mass with age than age-matched wild-type (WT) mice fed with either chow or high-fat diets. This phenomenon was attenuated by concomitant treatment with the antioxidants, melatonin or N-acetyl cysteine. Moreover, CKO mouse embryonic fibroblasts (MEFs) appeared to differentiate to adipocytes more easily than WT MEFs, showing increased H2O2 concentrations. Using 3T3-L1-derived adipocytes transfected with catalase-small interfering RNA, we confirmed that a more prominent lipogenesis occurred in catalase-deficient cells than in WT cells. Catalase-deficient adipocytes presented increased nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) expression but decreased adenosine monophosphate-activated protein kinase (AMPK) expression. Treatment with a NOX4 inhibitor or AMPK activator rescued the propensity for obesity of CKO mice. These findings suggest that excessive H2O2 and related oxidative stress increase body fat mass via both adipogenesis and lipogenesis. Manipulating NOX4 and AMPK in white adipocytes may be a therapeutic tool against obesity augmented by oxidative stress.


Subject(s)
Adipocytes, White , Hydrogen Peroxide , 3T3-L1 Cells , Adipogenesis , Animals , Catalase/genetics , Diet, High-Fat , Fibroblasts , Hyperplasia , Hypertrophy , Mice , Mice, Inbred C57BL , Obesity/genetics , Oxidative Stress
14.
Exp Mol Med ; 52(2): 238-252, 2020 02.
Article in English | MEDLINE | ID: mdl-32015410

ABSTRACT

Isocitrate dehydrogenase 2 (IDH2) is an NADP+-dependent enzyme that catalyzes the oxidative decarboxylation of isocitrate to α-ketoglutarate in the mitochondrial matrix, and is critical for the production of NADPH to limit the accumulation of mitochondrial reactive oxygen species (ROS). Here, we showed that high-fat diet (HFD) feeding resulted in accelerated weight gain in the IDH2KO mice due to a reduction in whole-body energy expenditure. Moreover, the levels of NADP+, NADPH, NAD+, and NADH were significantly decreased in the brown adipose tissue (BAT) of the HFD-fed IDH2KO animals, accompanied by decreased mitochondrial function and reduced expression of key genes involved in mitochondrial biogenesis, energy expenditure, and ROS resolution. Interestingly, these changes were partially reversed when the antioxidant butylated hydroxyanisole was added to the HFD. These observations reveal a crucial role for IDH2 in limiting ROS-dependent mitochondrial damage when BAT metabolism is normally enhanced to limit weight gain in response to dietary caloric overload.


Subject(s)
Adipose Tissue, Brown/metabolism , Isocitrate Dehydrogenase/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Stress, Physiological/physiology , Animals , Antioxidants/metabolism , Diet, High-Fat/adverse effects , Male , Mice , Mice, Inbred C57BL , NADP/metabolism , Organelle Biogenesis , Oxidation-Reduction , Reactive Oxygen Species/metabolism
15.
Inflamm Res ; 69(2): 191-202, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31897507

ABSTRACT

OBJECTIVES: Obesity is attributable to high free fatty acids, ER stress, oxidative stress and inflammation. The expression of IL-33, IL-1RL1 and IL-1RAP gene was observed in human visceral white fats, pre-adipocytes and adipocytes. The aim of this study was to determine whether IL1RAP and IL1RL1 gene variants were associated with obesity and inflammation mediators. METHODS: 3 SNPs of IL1RAP (rs9990107, rs3836449 and rs9290936) and 11 SNPs of IL1RL1 (rs3771180, rs13431828, rs3214363, rs1420101, rs12905, rs3771175, rs3821204, rs12712142, rs10204137, rs4988958, and rs10206753) were genotyped for 175 obesity (BMI ≥ 25) and 358 non-obesity (BMI < 25.0) subjects. The genotype of SNPs was determined by the Axiom Genome-Wide Human Assay. RESULTS: The allele and genotype frequencies of 2 SNPs in the IL1RAP gene (rs9990107 and rs3836449) and 11 SNPs in the IL1RL1 gene (rs3771180, rs13431828, rs3214363, rs1420101, rs12905, rs3771175, rs3821204, rs12712142, rs10204137, rs4988958 and rs10206753) were significantly associated between the obesity and non-obesity groups. The two haplotypes (GCTTATGAATT and TT-CGACCGCC) in block1 were associated with obesity. In the non-obesity group, genotype frequencies of rs3771180, rs13431828, rs3214363, rs10204137, rs4988958 and rs10206753 SNPs of IL1RL1 showed significant differences in the dominant models in lymphatic cell percentage. The genotype frequencies of rs1420101, rs21905, rs3821024 and rs12712142 SNPs of IL1RL1 showed significant differences in the dominant models in eosinophil percentage. CONCLUSIONS: Our results suggest that IL1RAP and IL1RL1 gene polymorphisms may be associated with obesity and inflammation mediators.


Subject(s)
Inflammation Mediators , Interleukin-1 Receptor Accessory Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/genetics , Obesity/genetics , Adult , Aged , Body Mass Index , Female , Gene Frequency , Genome-Wide Association Study , Genotype , Haplotypes , Humans , Lymphocytes , Male , Middle Aged , Polymorphism, Genetic/genetics , Polymorphism, Single Nucleotide
16.
Pflugers Arch ; 471(11-12): 1407-1418, 2019 12.
Article in English | MEDLINE | ID: mdl-31667577

ABSTRACT

Orexin A (OXA) is a neuropeptide associated with plasma insulin and leptin levels involved in body weight and appetite regulation. However, little is known about the effect of OXA on leptin secretion in adipocytes and its physiological roles. Leptin secretion and expression were analysed in 3T3-L1 adipocytes. Plasma leptin, adiponectin and insulin levels were measured by ELISA assay. Phosphorylated signal transducer and activator of transcription 3 (pSTAT3) levels in the hypothalamus were evaluated by western blotting. OXA dose-dependently suppressed leptin secretion from 3T3-L1 adipocytes by inhibiting its gene expression while facilitating adiponectin secretion. The leptin inhibition by OXA was mediated via orexin receptors (OXR1 and OXR2). In addition to the pathway via extracellular signal-regulated kinases, OXA triggered adenylyl cyclase-induced cAMP elevation, which results in protein kinase A-mediated activation of cAMP response element-binding proteins (CREB). Accordingly, CREB inhibition restored the OXA-induced downregulation of leptin gene expression and secretion. Exogenous OXA for 4 weeks decreased fasting plasma leptin levels and increased hypothalamic pSTAT3 levels in high-fat diet-fed mice, regardless of increase in body weight and food intake. These results suggest that high dose of OXA directly inhibits leptin mRNA expression and thus secretion in adipocytes, which may be a peripheral mechanism of OXA for its role in appetite drive during fasting. It may be also critical for lowering basal plasma leptin levels and thus maintaining postprandial hypothalamic leptin sensitivity.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Leptin/blood , Leptin/metabolism , Orexins/pharmacology , 3T3-L1 Cells , Animals , Appetite/drug effects , Body Weight/drug effects , Cell Line , Diet, High-Fat , Male , Mice , Mice, Inbred C57BL , Neuropeptides/metabolism , Orexin Receptors/metabolism
17.
PLoS One ; 14(9): e0222032, 2019.
Article in English | MEDLINE | ID: mdl-31504048

ABSTRACT

Type 2 diabetes is a metabolic disease with a group of metabolic derangements and inflammatory reactants in the serum. Despite the substantial public health implications, markers of diabetes progression with abdominal obesity are still needed to facilitate early detection and treatment. In this study, we performed a proteomic approach to identify differential target proteins underlying diabetes progression in patients with abdominal obesity. Proteomic differences were investigated in the serum of controls and patients with prediabetes or diabetes with or without abdominal obesity by 2-DE combined with MALDI-TOF-MS. Proteomics data were validated by western blot analyses and major protein-protein interactions were assessed using a network analysis with String database. Among 245 matched protein spots, 36 exhibited marked differences in normal patients with abdominal obesity, prediabetes, and diabetes compared to levels in normal patients without abdominal obesity. Seven (Alpha-1-antichymotrypsin, Alpha-1-antitrypsin, Apolipoprotein A-I, haptoglobin, retinol-binding protein 4, transthyretin, and zinc-alpha2-glycoprotein) of these spots exhibited significant differences between normal and prediabetes/diabetes patients. After a network analysis, functional annotation using Gene Ontology indicated that most of the identified proteins were involved in lipid transport, lipid localization, and the regulation of serum lipoprotein particle levels. Our results indicated that variation in the levels of these identified protein biomarkers has been reported in normal, prediabetes and diabetic Assessment of the levels of these biomarkers may contribute to the development of biomarkers for not only early diagnosis but also in prognosis of diabetes mellitus type 2.


Subject(s)
Diabetes Mellitus/blood , Obesity, Abdominal/complications , Proteome/chemistry , Adipokines , Adult , Apolipoproteins/blood , Biomarkers/blood , Carrier Proteins/blood , Diabetes Mellitus/diagnosis , Female , Glycoproteins/blood , Haptoglobins/analysis , Humans , Male , Middle Aged , Prealbumin/analysis , Republic of Korea , Retinol-Binding Proteins/analysis , alpha 1-Antitrypsin/blood
18.
Biosens Bioelectron ; 142: 111482, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31279172

ABSTRACT

In this study, we demonstrate a novel affinity peptide-incorporated electrochemical biosensor for the detection of acute kidney injury and the diabetic biomarker neutrophil gelatinase-associated lipocalin (NGAL). Biopanning of the M13 phage display library over immobilized NGAL led to the rapid identification of unique affinity peptide with an amino acid sequence of DRWVARDPASIF, and the peptide-displayed phage particles were found to be specific affinities for NGAL. To address the development of peptide-based electrochemical sensor, a series of synthetic peptides away from phage particles was rationally designed, chemically synthesized, and immobilized to a gold sensor layer. Among five synthetic peptide derivatives tested, NGAL BP1 was selected as most promising recognition receptor, and its binding affinity was monitored by SWV and EIS. Using EIS, the limit of detection (LOD) was 1.74 ng/mL, while SWV had a LOD of 3.93 ng/mL. The detection performance of the peptide-incorporated sensor was comparable to commercially available ELISA NGAL detection kits. In addition, the validation of the peptide sensor was also confirmed with plasma from patients, and it was observed that the sensitivity of the peptide sensor showed a statistically significant difference. Our results show that the phage and peptide sensor system could detect NGAL with high sensitivity and selectivity, and this suggests its potential use as a biosensing platform for monitoring NGAL in a miniaturized electrochemical biosensor.


Subject(s)
Biosensing Techniques/methods , Lipocalin-2/blood , Peptides/chemistry , Acute Kidney Injury/blood , Amino Acid Sequence , Binding Sites , Biomarkers/blood , Electrochemical Techniques/methods , Gold/chemistry , Humans , Limit of Detection , Lipocalin-2/analysis , Peptide Library
19.
Pflugers Arch ; 471(7): 1041, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31111222

ABSTRACT

The publisher would like to apologize for the failed cross-linking of the following Commentary by Jae-Hyung Park and Dae-Kyu Song.

20.
Leukemia ; 33(12): 2912-2923, 2019 12.
Article in English | MEDLINE | ID: mdl-31138843

ABSTRACT

A large body of evidence suggests that B-cell lymphomas with enhanced Myc expression are associated with an aggressive phenotype and poor prognosis, which makes Myc a compelling therapeutic target. Phosphodiesterase 4B (PDE4B), a main hydrolyzer of cyclic AMP (cAMP) in B cells, was shown to be involved in cell survival and drug resistance in diffuse large B cell lymphomas (DLBCL). However, the interrelationship between Myc and PDE4B remains unclear. Here, we first demonstrate the presence of the Myc-PDE4B feed-forward loop, in which Myc and PDE4B mutually reinforce the expression of each other. Next, the combined targeting of Myc and PDE4 synergistically prevented the proliferation and survival of B lymphoma cells in vitro and in a mouse xenograft model. We finally recapitulated this combinatorial effect in Eµ-myc transgenic mice; co-inhibition of Myc and PDE4 suppressed lymphomagenesis and restored B cell development to the wild type level that was associated with marked reduction in Myc levels, unveiling the critical role of the Myc-PDE4B amplification loop in the regulation of Myc expression and the pathogenesis of B cell lymphoma. These findings suggest that the disruption of the Myc-PDE4B circuitry can be exploited in the treatment of B cell malignancies.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/mortality , Proto-Oncogene Proteins c-myc/genetics , Animals , Biomarkers, Tumor , Cell Line, Tumor , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Disease Models, Animal , Humans , Immunohistochemistry , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/mortality , Lymphoma, Large B-Cell, Diffuse/pathology , Mice, Transgenic , Prognosis , Protein Binding , Proto-Oncogene Proteins c-myc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...