Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 600
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731926

ABSTRACT

The escalating prevalence of diabetes mellitus underscores the need for a comprehensive understanding of pancreatic beta cell function. Interest in glucose effectiveness has prompted the exploration of novel regulatory factors. The myeloid/lymphoid or mixed-lineage leukaemia gene (MLL) is widely recognised for its role in leukemogenesis and nuclear regulatory mechanisms through its histone methyltransferase activity in active chromatin. However, its function within pancreatic endocrine tissues remains elusive. Herein, we unveil a novel role of MLL in glucose metabolism and insulin secretion. MLL knockdown in ßHC-9 pancreatic beta cells diminished insulin secretion in response to glucose loading, paralleled by the downregulation of the glucose-sensitive genes SLC2a1 and SLC2a2. Similar observations were made in MLL heterozygous knockout mice (MLL+/-), which exhibited impaired glucose tolerance and reduced insulin secretion without morphological anomalies in pancreatic endocrine cells. The reduction in insulin secretion was independent of changes in beta cell mass or insulin granule morphology, suggesting the regulatory role of MLL in glucose-sensitive gene expression. The current results suggest that MLL interacts with circadian-related complexes to modulate the expression of glucose transporter genes, thereby regulating glucose sensing and insulin secretion. Our findings shed light on insulin secretion control, providing potential avenues for therapeutics against diabetes.


Subject(s)
Glucose Transporter Type 2 , Glucose , Histone-Lysine N-Methyltransferase , Insulin Secretion , Insulin-Secreting Cells , Myeloid-Lymphoid Leukemia Protein , Animals , Insulin-Secreting Cells/metabolism , Glucose/metabolism , Mice , Myeloid-Lymphoid Leukemia Protein/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 2/genetics , Gene Expression Regulation , Mice, Knockout , Insulin/metabolism , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 1/genetics , Cell Line , Male
2.
J Anim Sci ; 1022024 Jan 03.
Article in English | MEDLINE | ID: mdl-38619320

ABSTRACT

The present study aimed to investigate the effects of deoxynivalenol (DON) stimulation on inflammatory injury and the expression of the glucose transporters sodium-dependent glucose transporter 1 (SGLT1) and glucose transporter protein 2 (GLU2) in porcine small intestinal epithelial cells (IPEC-J2). Additionally, the study aimed to provide initial insights into the connection between the expression of glucose transporters and the inflammatory injury of IPEC-J2 cells. DON concentration and DON treatment time were determined using the CCK­8 assay. Accordingly, 1.0 µg/mL DON and treatment for 24 h were chosen for subsequent experiments. Then IPEC-J2 cells were treated without DON (CON, N = 6) or with 1 µg/mL DON (DON, N = 6). Lactate dehydrogenase (LDH) content, apoptosis rate, and proinflammatory cytokines including interleukin (IL)-1ß, Il-6, and tumor necrosis factor α (TNF-α) were measured. Additionally, the expression of AMP-activated protein kinase α1 (AMPK-α1), the content of glucose, intestinal alkaline phosphatase (AKP), and sodium/potassium-transporting adenosine triphosphatase (Na+/K+-ATPase) activity, and the expression of SGLT1 and GLU2 of IPEC-J2 cells were also analyzed. The results showed that DON exposure significantly increased LDH release and apoptosis rate of IPEC-J2 cells. Stimulation with DON resulted in significant cellular inflammatory damage, as evidenced by a significant increase in proinflammatory cytokines (IL-1ß, IL-6, and TNF-α). Additionally, DON caused damage to the glucose absorption capacity of IPEC-J2 cells, indicated by decreased levels of glucose content, AKP activity, Na+/K+-ATPase activity, AMPK-α1 protein expression, and SGLT1 expression. Correlation analysis revealed that glucose absorption capacity was negatively correlated with cell inflammatory cytokines. Based on the findings of this study, it can be preliminarily concluded that the cell inflammatory damage caused by DON may be associated with decreased glucose absorption.


Glucose is one of the most basic nutrients necessary to sustain animal life and plays a crucial role in animal body composition and energy metabolism. Previous studies suggested a link between glucose absorption and inflammatory injury. In the present study, deoxynivalenol (DON) stimulation caused severe inflammatory injury and reduced the glucose absorption capacity of IPEC-J2 cells. Pearson's correlation analysis revealed a negative correlation between glucose absorption capacity and cell inflammatory cytokines. Ultimately, it can be speculated that the cellular inflammatory response triggered by DON may be related to the altered expression of glucose transporters.


Subject(s)
Epithelial Cells , Glucose , Intestine, Small , Sodium-Glucose Transporter 1 , Trichothecenes , Animals , Trichothecenes/toxicity , Swine , Glucose/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 1/genetics , Cell Line , Intestine, Small/drug effects , Inflammation/chemically induced , Cytokines/metabolism , Cytokines/genetics , Biological Transport/drug effects , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 2/genetics , Apoptosis/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism
3.
Mol Biol Rep ; 50(8): 6963-6974, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37358764

ABSTRACT

The glucose transporter family has an important role in the initial stage of glucose metabolism; Glucose transporters 2 (GLUTs, encoded by the solute carrier family 2, SLC2A genes) is the major glucose transporter in ß-cells of pancreatic islets and hepatocytes but is also expressed in the small intestine, kidneys, and central nervous system; GLUT2 has a relatively low affinity to glucose. Under physiological conditions, GLUT2 transports glucose into cells and allows the glucose concentration to reach balance on the bilateral sides of the cellular membrane; Variation of GLUT2 is associated with various endocrine and metabolic disorders; In this study, we discussed the role of GLUT2 in participating in glucose metabolism and regulation in multiple organs and tissues and its effects on maintaining glucose homeostasis.


Subject(s)
Glucose , Islets of Langerhans , Glucose/metabolism , Islets of Langerhans/metabolism , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Hepatocytes/metabolism , Biological Transport , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism
4.
Biomolecules ; 13(3)2023 03 16.
Article in English | MEDLINE | ID: mdl-36979475

ABSTRACT

OBJECTIVE: Glucose transporter 2 (GLUT2) is expressed in the pancreatic ß-cell, intestine, liver, and kidney in mice. Although GLUT2 is considered as a major regulator of insulin secretion, in vivo contribution of ß-cell Glut2 to glucose-stimulated insulin secretion and systemic glucose homeostasis is undefined. Therefore, the main objective of this study is to determine the role of ß-cell Glut2 in regulating insulin secretion and blood glucose levels in mice. METHODS: We produced mice in which we can knock down Glut2 at a desired time specifically in ß-cells (ß-Glut2 KD) by crossing Glut2LoxP/LoxP mice with Ins1CreERT2 mouse strain and using the Cre-Lox recombination technique. We measured fasting blood glucose levels, glucose tolerance, and glucose-stimulated insulin secretion in the ß-Glut2 KD mice. We used qRT-PCR and immunofluorescence to validate the deficiency of ß-cell Glut2 in ß-Glut2 KD mice. RESULTS: We report that both male and female ß-Glut2 KD mice have normal glucose-stimulated insulin secretion. Moreover, the ß-Glut2 KD mice exhibit normal fasting blood glucose levels and glucose tolerance. The ß-Glut2 KD mice have upregulated GLUT1 in islets. CONCLUSIONS: Our findings demonstrate that normal ß-cell Glut2 expression is not essential for regulating glucose-stimulated insulin secretion and systemic glucose homeostasis in mice. Therefore, the currently assumed role of ß-cell GLUT2 in regulating insulin secretion and blood glucose levels needs to be recalibrated. This will allow an opportunity to determine the contribution of other ß-cell glucose transporters or factors whose normal expression may be necessary for mediating glucose stimulated insulin secretion.


Subject(s)
Glucose Transporter Type 2 , Insulin-Secreting Cells , Animals , Female , Male , Mice , Blood Glucose/metabolism , Glucose/pharmacology , Glucose/metabolism , Homeostasis , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Glucose Transporter Type 2/metabolism
5.
Sci Rep ; 12(1): 17717, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36271117

ABSTRACT

Feeding behavior is a complex process that depends on the ability of the brain to integrate hormonal and nutritional signals, such as glucose. One glucosensing mechanism relies on the glucose transporter 2 (GLUT2) in the hypothalamus, especially in radial glia-like cells called tanycytes. Here, we analyzed whether a GLUT2-dependent glucosensing mechanism is required for the normal regulation of feeding behavior in GFAP-positive tanycytes. Genetic inactivation of Glut2 in GFAP-expressing tanycytes was performed using Cre/Lox technology. The efficiency of GFAP-tanycyte targeting was analyzed in the anteroposterior and dorsoventral axes by evaluating GFP fluorescence. Feeding behavior, hormonal levels, neuronal activity using c-Fos, and neuropeptide expression were also analyzed in the fasting-to-refeeding transition. In basal conditions, Glut2-inactivated mice had normal food intake and meal patterns. Implementation of a preceeding fasting period led to decreased total food intake and a delay in meal initiation during refeeding. Additionally, Glut2 inactivation increased the number of c-Fos-positive cells in the ventromedial nucleus in response to fasting and a deregulation of Pomc expression in the fasting-to-refeeding transition. Thus, a GLUT2-dependent glucose-sensing mechanism in GFAP-tanycytes is required to control food consumption and promote meal initiation after a fasting period.


Subject(s)
Ependymoglial Cells , Feeding Behavior , Glucose Transporter Type 2 , Animals , Mice , Ependymoglial Cells/metabolism , Fasting , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glucose/metabolism , Hypothalamus/metabolism , Neuropeptides/metabolism , Pro-Opiomelanocortin/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Glucose Transporter Type 2/metabolism
6.
Hormones (Athens) ; 21(4): 625-640, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35843978

ABSTRACT

PURPOSE: Growing evidence has demonstrated that adversity in early life, especially in the prenatal and postnatal period, may change the programming of numerous body systems and cause the incidence of various disorders in later life. Accordingly, this experimental animal study aimed to investigate the effect of stress exposure during perinatal (prenatal and/or postnatal) on the induction of oxidative stress in the pancreas and its effect on glucose metabolism in adult rat offspring. METHODS: In this experimental study based on maternal exposure to variable stress throughout the perinatal period, the pups were divided into eight groups, as follows: control group (C); prepregnancy, pregnancy, lactation stress group (PPPLS); prepregnancy stress group (PPS); pregnancy stress group (PS); lactation stress group (LS); prepregnancy, pregnancy stress group (PPPS); pregnancy, lactation stress group (PLS); and prepregnancy, lactation stress group (PPLS). Following an overnight fast on postnatal day (PND) 64, plasma glucose, insulin, leptin levels, and lipid profiles were evaluated in the offspring groups. GLUT-2 protein levels, lipid peroxidation, antioxidant status, and number of beta-cells in the pancreatic islets of Langerhans as well as the weights of intra-abdominal fat and adrenal glands were assessed. Levels of plasma corticosterone were determined in the different groups of mothers and offspring. RESULTS: The levels of plasma corticosterone, insulin, and HOMA-B index increased, whereas glucose level and QUICKI index were reduced in the perinatal stress groups compared to C group (p < 0.001 to p < 0.05). Plasma triglyceride, LDL, and cholesterol level rose significantly, but HDL level decreased in the perinatal stress groups compared to C group (p < 0.001 to p < 0.05). Perinatal stress raised MDA concentrations and reduced the activities of antioxidant enzymes in plasma and pancreas compared to C group (p < 0.001 to p < 0.05). GLUT-2 protein levels and number of beta-cells in the stress groups declined compared to C group (p < 0.001 to p < 0.05). Intra-abdominal fat weight decreased in the PPS, PS, and LS groups compared to C group (p < 0.001 to p < 0.01), but adrenal gland weight remained unchanged. CONCLUSION: Our results showed that long-term exposure to elevated levels of corticosterone during critical development induces metabolic syndrome in adult male rats.


Subject(s)
Glucose Transporter Type 2 , Metabolic Diseases , Oxidative Stress , Prenatal Exposure Delayed Effects , Animals , Female , Male , Pregnancy , Rats , Antioxidants/metabolism , Corticosterone , Glucose Transporter Type 2/metabolism , Insulin , Lactation/metabolism , Rats, Wistar
7.
Sci Rep ; 12(1): 1429, 2022 01 26.
Article in English | MEDLINE | ID: mdl-35082341

ABSTRACT

The passive transport of glucose and related hexoses in human cells is facilitated by members of the glucose transporter family (GLUT, SLC2 gene family). GLUT3 is a high-affinity glucose transporter primarily responsible for glucose entry in neurons. Changes in its expression have been implicated in neurodegenerative diseases and cancer. GLUT3 inhibitors can provide new ways to probe the pathophysiological role of GLUT3 and tackle GLUT3-dependent cancers. Through in silico screening of an ~ 8 million compounds library against the inward- and outward-facing models of GLUT3, we selected ~ 200 ligand candidates. These were tested for in vivo inhibition of GLUT3 expressed in hexose transporter-deficient yeast cells, resulting in six new GLUT3 inhibitors. Examining their specificity for GLUT1-5 revealed that the most potent GLUT3 inhibitor (G3iA, IC50 ~ 7 µM) was most selective for GLUT3, inhibiting less strongly only GLUT2 (IC50 ~ 29 µM). None of the GLUT3 inhibitors affected GLUT5, three inhibited GLUT1 with equal or twofold lower potency, and four showed comparable or two- to fivefold better inhibition of GLUT4. G3iD was a pan-Class 1 GLUT inhibitor with the highest preference for GLUT4 (IC50 ~ 3.9 µM). Given the prevalence of GLUT1 and GLUT3 overexpression in many cancers and multiple myeloma's reliance on GLUT4, these GLUT3 inhibitors may discriminately hinder glucose entry into various cancer cells, promising novel therapeutic avenues in oncology.


Subject(s)
Drug Discovery , Glucose Transporter Type 3/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/drug effects , Small Molecule Libraries/pharmacology , Binding Sites , Biological Transport/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/chemistry , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 2/antagonists & inhibitors , Glucose Transporter Type 2/chemistry , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 3/antagonists & inhibitors , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Glucose Transporter Type 4/antagonists & inhibitors , Glucose Transporter Type 4/chemistry , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Glucose Transporter Type 5/antagonists & inhibitors , Glucose Transporter Type 5/chemistry , Glucose Transporter Type 5/genetics , Glucose Transporter Type 5/metabolism , Heterocyclic Compounds, 3-Ring/chemistry , High-Throughput Screening Assays , Humans , Models, Molecular , Neoplasms/drug therapy , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Small Molecule Libraries/chemistry
8.
Life Sci ; 290: 120261, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34968468

ABSTRACT

AIMS: We previously showed that hindlimb ischemia-reperfusion (IR) enhanced glucose uptake in the liver through the activation of the parasympathetic nervous system. Although we suggested that the key glucose transporter (GLUT) in this hepatic glucose uptake was GLUT4 by western blotting, the molecular weight of GLUT4 was nearly the same as that of GLUT2, which is predominantly expressed in the liver. We primarily conducted a histological evaluation to determine whether IR specifically accelerates the overexpression of GLUT4, rather than GLUT2, in the hepatocytes in vitro and in vivo. MAIN METHODS: A total of 54 male C57BL/6J mice were used and subjected to 3 min hindlimb ischemia repeated three times with 3 min interval. Focusing on the area connecting portal and central veins, the GLUT4 and GLUT2 expression in the hepatocytes were examined by real-time PCR and immunohistochemically. Moreover, the alteration of GLUT4 and GLUT2 expression by acetylcholine in the primary hepatocytes were examined by immunofluorescence. KEY FINDINGS: IR significantly upregulated the GLUT4, rather than GLUT2, expression in both mRNA and protein in the liver. Histological examination revealed marked glycogen storage in zone1, the periportal area, coincident with the enhanced GLUT4 immunoreactivity, in the IR-treated liver. Incubation of primary hepatocytes with acetylcholine induced the appearance of GLUT4 on the membrane peripheries. SIGNIFICANCE: The overexpression of GLUT4 on the membrane peripheries contributed to increasing glucose uptake found in IR-treated livers. This acceleration of glucose uptake via GLUT4 may induce marked glycogen storage in zone1 through energy production linked with increased glucose preference.


Subject(s)
Glucose Transporter Type 4/metabolism , Glycogen/metabolism , Ischemic Preconditioning/methods , Animals , Cell Membrane/metabolism , Gene Expression/genetics , Gene Expression Regulation/genetics , Glucose/metabolism , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 4/genetics , Hepatocytes/metabolism , Insulin/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Reperfusion Injury/metabolism
9.
J Neurochem ; 160(2): 283-296, 2022 01.
Article in English | MEDLINE | ID: mdl-34726780

ABSTRACT

The retina has the highest relative energy consumption of any tissue, depending on a steady supply of glucose from the bloodstream. Glucose uptake is mediated by specific transporters whose regulation and expression are critical for the pathogenesis of many diseases, including diabetes and diabetic retinopathy. Here, we used immunofluorescence to show that glucose transporter-2 (GLUT2) is expressed in horizontal cells of the mouse neuroretina in proximity to inner retinal capillaries. To study the function of GLUT2 in the murine retina, we used organotypic retinal explants, cultivated under entirely controlled, serum-free conditions and exposed them to streptozotocin, a cytotoxic drug transported exclusively by GLUT2. Contrary to our expectations, streptozotocin did not measurably affect horizontal cell viability, while it ablated rod and cone photoreceptors in a concentration-dependent manner. Staining for poly-ADP-ribose (PAR) indicated that the detrimental effect of streptozotocin on photoreceptors may be associated with DNA damage. The negative effect of streptozotocin on the viability of rod photoreceptors was counteracted by co-administration of either the inhibitor of connexin-formed hemi-channels meclofenamic acid or the blocker of clathrin-mediated endocytosis dynasore. Remarkably, cone photoreceptors were not protected from streptozotocin-induced degeneration by neither of the two drugs. Overall, these data suggest the existence of a GLUT2-dependent glucose transport shuttle, from horizontal cells into photoreceptor synapses. Moreover, our study points at different glucose uptake mechanisms in rod and cone photoreceptors.


Subject(s)
Glucose Transporter Type 2/metabolism , Glucose/metabolism , Photoreceptor Cells/metabolism , Retinal Horizontal Cells/metabolism , Synapses/metabolism , Animals , Biological Transport , Mice , Retina/metabolism
10.
Genes (Basel) ; 12(11)2021 11 10.
Article in English | MEDLINE | ID: mdl-34828390

ABSTRACT

Fanconi-Bickel syndrome (FBS) is a very rare but distinct clinical entity with the combined features of hepatic glycogen storage disease, generalized proximal renal tubular dysfunction with disproportionately severe glucosuria, and impaired galactose tolerance. Here, we report five cases (out of 93 diagnosed in our lab) with pathogenic variants on both GLUT2 (SLC2A2) alleles. They come from 3 families and presented with an exceptionally mild clinical course. This course was correlated to data from old and most recent expression and transport studies in Xenopus oocytes. GLUT2 genotype in patients 1 and 2 was p.[153_4delLI];[P417R] with the first variant exhibiting normal membrane expression and partially retained transport activity (5.8%) for 2-deoxyglucose. In patient 3, the very first GLUT2 variant ever detected (p.V197I) was found, but for the first time it was present in a patient in the homozygous state. This variant had also shown unaffected membrane expression and remarkable residual activity (8%). The genotype in patient 4, p.[153_4delLI];[(E440A)], again included the 2-amino-acid deletion with residual transporter function, and patient 5 is the first found to be homozygous for this variant. Our results provide further evidence for a genotype-phenotype correlation in patients with GLUT2 variants; non-functional variants result in the full picture of FBS while dysfunctional variants may result in milder presentations, even glucosuria only, without other typical signs of FBS.


Subject(s)
Fanconi Syndrome/genetics , Glucose Transporter Type 2/genetics , Mutation , Phenotype , Adolescent , Adult , Animals , Fanconi Syndrome/pathology , Female , Genotype , Glucose/metabolism , Glucose Transporter Type 2/metabolism , Homozygote , Humans , Infant , Male , Pedigree , Xenopus
11.
Dis Markers ; 2021: 2821673, 2021.
Article in English | MEDLINE | ID: mdl-34745386

ABSTRACT

To elucidate the mechanism of how the liver participates in PM2.5-caused insulin resistance. A novel Wistar rat model was developed in this study by instilling a suspension of lyophilized PM2.5 sample (2.5 mg/kg, 5 mg/kg, or 10 mg/kg) collected from the atmosphere. Systemic insulin resistance indicators, including serum fasting blood glucose (FBG), fasting insulin (FINS), Homeostatic Model Assessment for Insulin Resistance (HOMA-IR), and hemoglobin A1 (HbA1), were upregulated by the PM2.5 instillation. The area under the curve (AUCglu) calculated by intraperitoneal glucose tolerance testing (IPGTT) was also significantly greater in the PM2.5 instillation groups. Additionally, PM2.5 instillation was found to cause liver damage and inflammation. The serum levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), total bilirubin (TBIL), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) were significantly elevated by PM2.5 instillation. PM2.5 also triggered IL-6 and TNF-α transcription but inhibited mRNA synthesis and suppressed signaling activation of the insulin-phosphoinositide 3-kinase- (PI3K-) Akt-glucose transporter 2 (GLUT2) pathway in the rat liver by reducing the ratio of phosphorylated Akt to phosphorylated insulin receptor substrate 1 (IRS-1). Thus, PM2.5-induced inflammation activation and insulin signaling inhibition in the rat liver contribute to the development of systemic insulin resistance.


Subject(s)
Insulin Resistance , Liver/metabolism , Particulate Matter/toxicity , Signal Transduction , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Bilirubin/blood , Blood Glucose/metabolism , Glucose Transporter Type 2/metabolism , Hemoglobins/metabolism , Insulin/blood , Interleukin-6/blood , Liver/drug effects , Male , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/blood
12.
BMC Cancer ; 21(1): 1026, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34525987

ABSTRACT

BACKGROUND: Current treatment methods for patients with triple-negative breast cancer (TNBC) are very limited, and the prognosis of TNBC is relatively poor. It has been reported that glucose transporter 1 (GLUT1) is overexpressed in breast cancer cells; however, its association with the prognosis is mostly unclear. Moreover, retinoblastoma gene 1 (RB1) might be used as a biomarker for the sensitivity of breast cancer cells to GLUT1 inhibitors, which brought us to the hypothesis that there might be a close correlation between the expression of GLUT1-4 and the expression of RB1. METHODS: In this study, we systematically analyzed the co-expression of GLUT1-4 and the influence of GLUT1-4 gene expression on the prognosis of breast cancer using data mining methods. We also explored possible relationships between GLUT1-4 and RB1 expression in breast cancer tissues. We used public databases such as ONCOMINE, GEPIA, LinkedOmics, and COEXPEDIA. RESULTS: According to the results, the mRNA expression of SLC2A1 was significantly higher in breast cancer, while the expression levels of SLC2A2-4 were downregulated. The results also indicate that GLUT1 expression does not have significant influence on the overall survival of patients with breast cancer. The mRNA expression of SLC2A1 and RB1 is significantly correlated, which means that tissues with high RB1 mRNA expression might have relatively higher mRNA expression of SLC2A1; however, further study analyzing their roles in the expression regulation pathways with human samples is needed to verify the hypothesis. CONCLUSIONS: The mRNA expression of SLC2A1 was significantly higher in breast cancer. The overall survival of breast cancer patients wasn't significantly correlated with GLUT1-4 expression. The mRNA expression of SLC2A1 and RB1 is significantly correlated according to the analysis conducted in LinkedOmics. It provides reference for future possible individualized treatment of TNBC using GLUT1 inhibitors, especially in patients with higher mRNA expression of RB1. Further study analyzing the roles of these two genes in the regulation pathways is needed.


Subject(s)
Glucose Transporter Type 1/metabolism , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 3/metabolism , Glucose Transporter Type 4/metabolism , Retinoblastoma Binding Proteins/metabolism , Triple Negative Breast Neoplasms/genetics , Ubiquitin-Protein Ligases/metabolism , Databases, Genetic , Datasets as Topic , Down-Regulation , Female , Genes, Retinoblastoma , Glucose Transporter Type 1/genetics , Glucose Transporter Type 2/genetics , Glucose Transporter Type 3/genetics , Glucose Transporter Type 4/genetics , Humans , Prognosis , RNA, Messenger/metabolism , Retinoblastoma Binding Proteins/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/mortality , Ubiquitin-Protein Ligases/genetics , Up-Regulation
13.
Cells ; 10(8)2021 08 07.
Article in English | MEDLINE | ID: mdl-34440786

ABSTRACT

The production of functional islet-like cells from human-induced pluripotent stem cells (hiPSCs) is a promising strategy for the therapeutic use and disease modeling for type 1 diabetes. However, the production cost of islet-like cells is extremely high due to the use of expensive growth factors for differentiation. In a conventional culture method, growth factors and beneficial autocrine factors remaining in the culture medium are removed along with toxic metabolites during the medium change, and it limits the efficient utilization of those factors. In this study, we demonstrated that the dialysis suspension culture system is possible to reduce the usage of growth factors to one-third in the differentiation of hiPSC-derived endocrine progenitor cells to islet-like cells by reducing the medium change frequency with the refinement of the culture medium. Furthermore, the expression levels of hormone-secretion-related genes and the efficiency of differentiation were improved with the dialysis suspension culture system, possibly due to the retaining of autocrine factors. In addition, we confirmed several improvements required for the further study of the dialysis culture system. These findings showed the promising possibility of the dialysis suspension culture system for the low-cost production of islet-like cells.


Subject(s)
Cell Differentiation/drug effects , Culture Media/pharmacology , Dialysis Solutions/pharmacology , Induced Pluripotent Stem Cells/drug effects , Islets of Langerhans/drug effects , Renal Dialysis , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Aggregation/drug effects , Cell Aggregation/genetics , Cell Culture Techniques/methods , Cell Differentiation/genetics , Cell Line , Culture Media/chemistry , Dialysis Solutions/chemistry , Endocrine System/cytology , Endocrine System/drug effects , Endocrine System/metabolism , Gene Expression/drug effects , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism
14.
Nutrients ; 13(8)2021 Jul 26.
Article in English | MEDLINE | ID: mdl-34444712

ABSTRACT

Glucagon-like peptide 1 (GLP-1) and PAS kinase (PASK) control glucose and energy homeostasis according to nutritional status. Thus, both glucose availability and GLP-1 lead to hepatic glycogen synthesis or degradation. We used a murine model to discover whether PASK mediates the effect of exendin-4 (GLP-1 analogue) in the adaptation of hepatic glycogen metabolism to nutritional status. The results indicate that both exendin-4 and fasting block the Pask expression, and PASK deficiency disrupts the physiological levels of blood GLP1 and the expression of hepatic GLP1 receptors after fasting. Under a non-fasted state, exendin-4 treatment blocks AKT activation, whereby Glucokinase and Sterol Regulatory Element-Binding Protein-1c (Srebp1c) expressions were inhibited. Furthermore, the expression of certain lipogenic genes was impaired, while increasing Glucose Transporter 2 (GLUT2) and Glycogen Synthase (GYS). Moreover, exendin-4 treatment under fasted conditions avoided Glucose 6-Phosphatase (G6pase) expression, while maintaining high GYS and its activation state. These results lead to an abnormal glycogen accumulation in the liver under fasting, both in PASK-deficient mice and in exendin-4 treated wild-type mice. In short, exendin-4 and PASK both regulate glucose transport and glycogen storage, and some of the exendin-4 effects could therefore be due to the blocking of the Pask expression.


Subject(s)
Adaptation, Physiological , Fasting , Liver Glycogen/metabolism , Liver/metabolism , Nutritional Status , Protein Serine-Threonine Kinases/metabolism , Animals , Exenatide/metabolism , Exenatide/pharmacology , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Glucokinase/metabolism , Glucose/metabolism , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Up-Regulation , Weight Loss
15.
J Mol Endocrinol ; 67(3): 71-82, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34223824

ABSTRACT

Glucose transporter type 2 (GLUT2), encoded by the SLC2A2 gene, is an essential component of glucose-stimulated insulin secretion in pancreatic islet ß-cells. Like that of the gene encoding insulin, expression of the SLC2A2 gene expression is closely linked to ß-cell functionality in rodents, but the mechanism by which ß-cell-specific expression of SLC2A2 is controlled remains unclear. In this report, to identify putative enhancer elements of the mouse Slc2a2 gene, we examined evolutional conservation of the nucleotide sequence of its genomic locus, together with ChIP-seq data of histone modifications and various transcription factors published in previous studies. Using luciferase reporter assays, we found that an evolutionarily conserved region (ECR) located approximately 40 kbp downstream of the transcription start site of Slc2a2 functions as an active enhancer in the MIN6 ß-cell line. We also found that three ß-cell-enriched transcription factors, MafA, NeuroD1, and HNF1ß, synergistically activate transcription through this 3' downstream distal enhancer (ECR3') and the proximal promoter region of the gene. Our data also indicate that the simultaneous binding of HNF1ß to its target sites within the promoter and ECR3' of Slc2a2 is indispensable for transcriptional activation, and that binding of MafA and NeuroD1 to their respective target sites within the ECR3' enhances transcription. Co-immunoprecipitation experiments suggested that MafA, NeuroD1, and HNF1ß interact with each other. Overall, these results suggest that promoter-enhancer communication through MafA, NeuroD1, and HNF1ß is critical for Slc2a2 gene expression. These findings provide clues to help elucidate the mechanism of regulation of Slc2a2 gene expression in ß-cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation , Glucose Transporter Type 2/genetics , Hepatocyte Nuclear Factor 1-beta/metabolism , Insulin-Secreting Cells/metabolism , Maf Transcription Factors, Large/metabolism , Animals , Conserved Sequence , Enhancer Elements, Genetic , Glucose Transporter Type 2/metabolism , Mice , Promoter Regions, Genetic , Protein Binding , Response Elements , Transcriptional Activation
16.
Mar Drugs ; 19(5)2021 May 18.
Article in English | MEDLINE | ID: mdl-34069768

ABSTRACT

The Red Sea is one of the most biodiverse aquatic ecosystems. Notably, seagrasses possess a crucial ecological significance. Among them are the two taxa Halophila stipulacea (Forsk.) Aschers., and Thalassia hemprichii (Ehrenb. ex Solms) Asch., which were formally ranked together with the genus Enhalus in three separate families. Nevertheless, they have been recently classified as three subfamilies within Hydrocharitaceae. The interest of this study is to explore their metabolic profiles through ultra-high-performance liquid chromatography-high-resolution mass spectrometry (UPLC-HRMS/MS) analysis in synergism with molecular networking and to assess their chemosystematics relationship. A total of 144 metabolites were annotated, encompassing phenolic acids, flavonoids, terpenoids, and lipids. Furthermore, three new phenolic acids; methoxy benzoic acid-O-sulphate (16), O-caffeoyl-O-hydroxyl dimethoxy benzoyl tartaric acid (26), dimethoxy benzoic acid-O-sulphate (30), a new flavanone glycoside; hexahydroxy-monomethoxy flavanone-O-glucoside (28), and a new steviol glycoside; rebaudioside-O-acetate (96) were tentatively described. Additionally, the evaluation of the antidiabetic potential of both taxa displayed an inherited higher activity of H. stipulaceae in alleviating the oxidative stress and dyslipidemia associated with diabetes. Hence, the current research significantly suggested Halophila, Thalassia, and Enhalus categorization in three different taxonomic ranks based on their intergeneric and interspecific relationship among them and supported the consideration of seagrasses in natural antidiabetic studies.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Hydrocharitaceae/chemistry , Hypoglycemic Agents/pharmacology , Metabolome , Animals , Blood Glucose/drug effects , Chromatography, High Pressure Liquid , Enzyme Assays , Glucose Transporter Type 2/metabolism , Hydrocharitaceae/genetics , Hydrolysis , Hypoglycemic Agents/therapeutic use , Indian Ocean , Insulin/blood , Male , Malondialdehyde/metabolism , Mass Spectrometry , Nitric Oxide/blood , Phylogeny , Phytochemicals/analysis , Rats, Wistar
17.
Nat Commun ; 12(1): 3133, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34035238

ABSTRACT

Heterozygous HNF1A gene mutations can cause maturity onset diabetes of the young 3 (MODY3), characterized by insulin secretion defects. However, specific mechanisms of MODY3 in humans remain unclear due to lack of access to diseased human pancreatic cells. Here, we utilize MODY3 patient-derived human induced pluripotent stem cells (hiPSCs) to study the effect(s) of a causal HNF1A+/H126D mutation on pancreatic function. Molecular dynamics simulations predict that the H126D mutation could compromise DNA binding and gene target transcription. Genome-wide RNA-Seq and ChIP-Seq analyses on MODY3 hiPSC-derived endocrine progenitors reveal numerous HNF1A gene targets affected by the mutation. We find decreased glucose transporter GLUT2 expression, which is associated with reduced glucose uptake and ATP production in the MODY3 hiPSC-derived ß-like cells. Overall, our findings reveal the importance of HNF1A in regulating GLUT2 and several genes involved in insulin secretion that can account for the insulin secretory defect clinically observed in MODY3 patients.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Glucose Transporter Type 2/genetics , Glucose/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Insulin Secretion/genetics , Insulin-Secreting Cells/metabolism , Mutation , Cells, Cultured , Chromatin Immunoprecipitation Sequencing/methods , Diabetes Mellitus, Type 2/metabolism , Female , Glucose Transporter Type 2/metabolism , Hepatocyte Nuclear Factor 1-alpha/chemistry , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Insulin-Secreting Cells/cytology , Male , Molecular Dynamics Simulation , Pedigree , Protein Domains
18.
Mol Ther ; 29(9): 2854-2872, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33895325

ABSTRACT

The processing, maturation, and secretion of insulin are under precise regulation, and dysregulation causes profound defects in glucose handling, leading to diabetes. Tmem30a is the ß subunit of the phosphatidylserine (PS) flippase, which maintains the membrane asymmetric distribution of PS. Tmem30a regulates cell survival and the localization of subcellular structures and is thus critical to the normal function of multiple physiological systems. Here, we show that conditional knockout of Tmem30a specifically in pancreatic islet ß cells leads to obesity, hyperglycemia, glucose intolerance, hyperinsulinemia, and insulin resistance in mice, due to insufficient insulin release. Moreover, we reveal that Tmem30a plays an essential role in clathrin-mediated vesicle transport between the trans Golgi network (TGN) and the plasma membrane (PM), which comprises immature secretory granule (ISG) budding at the TGN. We also find that Tmem30a deficiency impairs clathrin-mediated vesicle budding and thus blocks both insulin maturation in ISGs and the transport of glucose-sensing Glut2 to the PM. Collectively, these disruptions compromise both insulin secretion and glucose sensitivity, thus contributing to impairments in glucose-stimulated insulin secretion. Taken together, our data demonstrate an important role of Tmem30a in insulin maturation and glucose metabolic homeostasis and suggest the importance of membrane phospholipid distribution in metabolic disorders.


Subject(s)
Glucose Intolerance/genetics , Glucose Transporter Type 2/metabolism , Hyperglycemia/genetics , Hyperinsulinism/genetics , Insulin Resistance/genetics , Insulin/metabolism , Membrane Proteins/genetics , Obesity/genetics , Animals , Cell Membrane/metabolism , Disease Models, Animal , Gene Knockout Techniques , Glucose/adverse effects , Glucose Intolerance/metabolism , Hyperglycemia/metabolism , Hyperinsulinism/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mice , Obesity/metabolism , Phosphatidylserines/metabolism , trans-Golgi Network/metabolism
19.
J Sci Food Agric ; 101(14): 5984-5991, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33856052

ABSTRACT

BACKGROUND: Nutritional modulations may be considered a strategy to protect mental health. Neuronal homeostasis is highly dependent on the availability of glucose, which represents the primary energy source for the brain. In this study, we evaluated the effects of walnut intake and fructose-rich diet on the expression of glucose transporters (GLUTs) in two rat brain regions: hypothalamus and hippocampus. RESULTS: Our results show that walnut supplementation of fructose-fed animals restored the hypothalamic content of GLUT1 and GLUT3 protein. Furthermore, walnut intake did not affect increased hypothalamic GLUT2 content upon fructose consumption. These effects were accompanied by distinctive alterations of hippocampal GLUTs levels. Specifically, walnut intake increased GLUT1 content, whereas GLUT2 protein was decreased within the rat hippocampus after both individual and combined treatments. CONCLUSION: Overall, our study suggests that walnut supplementation exerted modulatory effects on the glucose transporters within specific brain regions in the presence of developed metabolic disorder. © 2021 Society of Chemical Industry.


Subject(s)
Glucose Transporter Type 1/metabolism , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 3/metabolism , Hippocampus/metabolism , Hypothalamus/metabolism , Juglans/metabolism , Animals , Fructose/metabolism , Glucose/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 2/genetics , Glucose Transporter Type 3/genetics , Male , Nuts/metabolism , Rats , Rats, Wistar
20.
J Biochem Mol Toxicol ; 35(6): 1-12, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33724628

ABSTRACT

The current study was done to measure the role of piperlongumine (PL) on hyperglycemia interrelated oxidative stress-mediated inflammation and apoptosis, inflammatory stress, and the diabetic insulin receptor substrate 2 (IRS2), protein kinase B (AKT), and glucose transporter 2 (GLUT-2)/4 signaling pathway in streptozotocin (STZ)-persuaded diabetic animals. Diabetes was initiated in experimental animals via a single dose intraperitoneal inoculation of STZ. Diabetic rats revealed an augmented blood-glucose level with drastically diminished plasma-insulin status. The functions of antioxidants were diminished with enhanced lipid peroxidation, conjugated dienes, and protein carbonyls noticed in diabetic rats'  plasma and pancreatic tissues. An elevation of nuclear factor-κB (NF-κB), tumor necrosis factor-α, and interleukin-6 proteins was noticed in pancreatic tissues as well as IRS2, AKT, GLUT-2, and GLUT-4 marker expressions were quantified in the hepatic tissue of control and diabetic rats. Oral administration of PL for 30 days drastically lowered glucose and higher insulin status in STZ-induced diabetic rats. Impressively, PL oral supplementation considerably restored the antioxidant levels and reduced inflammation and diabetic marker expressions in STZ-diabetic rats. These results were supported through a histological study. Moreover, PL also augmented the level of B-cell lymphoma 2 and diminished the level of Bcl-2-associated X protein in STZ-treated rat's hepatic tissues. Thus, we concluded that PL excellently rescued pancreatic ß cells through mitigating hyperglycemia via dynamic insulin secretion, activating antioxidants, and inhibiting inflammation and apoptosis in the pancreatic and hepatic tissue of diabetic rats.


Subject(s)
Apoptosis/drug effects , Diabetes Mellitus, Experimental , Dioxolanes/pharmacology , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 4/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Animals , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Male , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...