Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Biochem Biophys Res Commun ; 485(2): 328-334, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28232184

ABSTRACT

OBJECTIVES: To investigate whether the activation of pancreatic stellate cells (PSCs) leads to pancreatic ß-cell dysfunction in type 2 diabetes mellitus (T2DM). METHODS: The pancreases of Otsuka Long-Evans Tokushima Fatty (OLETF) rats, an animal model of T2DM, and patient with T2DM were analyzed. And the in vitro and in vivo effects of pirfenidone, an antifibrotic agent, on PSC activation, islet fibrosis, and ß-cells were studied. RESULTS: The extent of islet fibrosis and the percentage of activated PSCs, positive for α-smooth muscle actin, in the islets were significantly greater in OLETF rats compared with non-diabetic rats. Also, the extent of islet fibrosis in patients with T2DM was slightly greater compared with age- and BMI-matched non-diabetic patients. In rat PSCs cultured with high glucose for 72 h, pirfenidone produced decreases in cell proliferation, release of collagen, and the expression of fibronectin and connective tissue growth factor. Treatment of OLETF rats with pirfenidone for 16 weeks decreased the activation of PSCs and the extent of islet fibrosis, but did not enhance glucose tolerance, pancreatic insulin content, or ß-cell mass. CONCLUSIONS: Activated PSCs in islets might lead to islet fibrosis in T2DM. However, PSC activation itself might not contribute significantly to progressive ß-cell failure in T2DM.


Subject(s)
Diabetes Mellitus, Type 2/pathology , Insulin-Secreting Cells/pathology , Pancreas/pathology , Pancreatic Stellate Cells/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cells, Cultured , Diabetes Mellitus, Type 2/drug therapy , Fibrosis/pathology , Humans , Insulin-Secreting Cells/drug effects , Male , Pancreas/drug effects , Pancreatic Stellate Cells/drug effects , Pyridones/pharmacology , Pyridones/therapeutic use , Rats, Inbred OLETF , Rats, Sprague-Dawley
2.
Biochem Biophys Res Commun ; 478(3): 1080-6, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27524234

ABSTRACT

LONG HYPOCOTYL5 (HY5) and STF1 (Soybean TGACG-motif binding Factor 1) are two related bZIP transcription factors that play a positive role in photomorphogenesis and hormonal signaling. In this study, we compared full length STF1 and truncated STF1 overexpression lines and found that the C-terminal 133 amino acids (194-306) possess all the HY5-like function in Arabidopsis. The STF1-DC1 mutant (1-306), with a 20 amino acid deletion at the carboxy terminus, failed to complement the hy5 mutant phenotype, which suggests an intact C-terminus is required for STF1 function. To understand the role of the C-terminal domain in photomorphogenesis we used a yeast two-hybrid screen to isolate proteins that bind to the STF1 C-terminus. We isolated three soybean cDNAs encoding the zinc-finger proteins GmSTO, GmSTH, and GmSTH2, which interact with STF1. These proteins belong to a family of B-box zinc finger proteins that include Arabidopsis SALT TOLERANCE (STO) and STO HOMOLOG (STH) and STH2, which play a role in light-dependent development and gene expression. The C-terminal 63 amino acids of STF1, containing a leucine zipper and the two N-terminal B-boxes, contains the domain involved in interactions between STF1 and GmSTO. In addition, we identified an interaction between soybean COP1 (GmCOP1) and GmSTO and GmSTH, as well as STF1, which strongly suggests the presence of a similar regulatory circuit for light signaling in soybean as in Arabidopsis. This study shows that photomorphogenic control requires complex molecular interactions among several different classes of transcription factors such as bZIP, B-box factors, and COP1, a ubiquitin ligase.


Subject(s)
Glycine max/growth & development , Glycine max/radiation effects , Light , Plant Proteins/isolation & purification , Plant Proteins/metabolism , Protein Interaction Maps , Zinc Fingers , Arabidopsis/genetics , DNA, Complementary/isolation & purification , Genes, Reporter , Genetic Complementation Test , Mutation/genetics , Plant Proteins/chemistry , Plants, Genetically Modified , Protein Binding , Protein Domains , Protein Interaction Domains and Motifs , Protein Transport , Saccharomyces cerevisiae/metabolism , Glycine max/metabolism , Subcellular Fractions/metabolism , Nicotiana/genetics , Transcription, Genetic , Transcriptional Activation/genetics
3.
Biochem Biophys Res Commun ; 443(3): 1021-7, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24365150

ABSTRACT

Recent studies have demonstrated that adult cells such as pancreatic exocrine cells can be converted to pancreatic ß-cells in a process called cell reprogramming. Enteroendocrine cells and ß-cells share similar pathways of differentiation during embryonic development. Notably, enteroendocrine K cells express many of the key proteins found in ß-cells. Thus, K cells could be reprogrammed to ß-cells under certain conditions. However, there is no clear evidence on whether these cells convert to ß-cells. K cells were selected from STC-1 cells, an enteroendocrine cell line expressing multiple hormones. K cells were found to express many genes of transcription factors crucial for islet development and differentiation except for Nkx6.1 and Neurogenin3. A K cell clone stably expressing Nkx6.1 (Nkx6.1(+)-K cells) was established. Induction of Neurogenin3 expression in Nkx6.1(+)-K cells, by either treatment with a γ-secretase inhibitor or infection with a recombinant adenovirus expressing Neurogenin3, led to a significant increase in Insulin1 mRNA expression. After infection with the adenovirus expressing Neurogenin3 and reaggregation in suspension culture, about 50% of Nkx6.1(+)-K cells expressed insulin as determined by immunostaining. The intracellular insulin content was increased markedly. Electron microscopy revealed the presence of insulin granules. However, glucose-stimulated insulin secretion was defective, and there was no glucose lowering effect after transplantation of these cells in diabetic mice. In conclusion, we demonstrated that K cells could be reprogrammed partially to ß-cells through the combined expression of Nkx6.1 and Neurogenin3, and reaggregation in suspension culture.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Culture Techniques/methods , Cellular Reprogramming , Enteroendocrine Cells/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Nerve Tissue Proteins/metabolism , Animals , Cell Aggregation , Enteroendocrine Cells/cytology , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Insulin-Secreting Cells/ultrastructure , Mice , Mice, Nude , Rats , Suspensions
4.
J Cell Biochem ; 114(4): 844-53, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23097217

ABSTRACT

Glucagon-like peptide-1 (GLP-1) and its potent agonists have been widely studied in pancreatic islet ß-cells. However, GLP-1 receptors are present in many extrapancreatic tissues including macrophages, and thus GLP-1 may have diverse actions in these tissues and cells. Therefore, we examined the mechanism by which exendin-4 (EX-4), a potent GLP-1 receptor agonist, inhibits lipopolysaccharide (LPS)-induced iNOS expression in Raw264.7 macrophage cells. EX-4 significantly inhibited LPS-induced iNOS protein expression and nitrite production. However, Northern blot and promoter analyses demonstrated that EX-4 did not inhibit LPS-induced iNOS mRNA expression and iNOS promoter activity. Electrophoretic mobility shift assay (EMSA) showed that EX-4 did not alter the binding activity of NF-κB to the iNOS promoter. Consistent with the result of EMSA, LPS-induced IκBα phosphorylation and nuclear translocation of p65 were not inhibited by EX-4. Also, actinomycin D chase study and the promoter assay using the construct containing 3'-untranslated region of iNOS showed that EX-4 did not affect iNOS mRNA stability. Meanwhile, cycloheximide chase study demonstrated that EX-4 significantly accelerated iNOS protein degradation. The EX-4 inhibition of LPS-induced iNOS protein was significantly reversed by adenylate cyclase inhibitors (MDL-12330A and SQ 22536), a PKA inhibitor (H-89) and PKAα gene silencing. These findings suggest that EX-4 inhibited LPS-induced iNOS expression at protein level, but not at transcriptional mechanism of iNOS gene and this inhibitory effect of EX-4 was mainly dependent on cAMP/PKA system.


Subject(s)
Cyclic AMP/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Nitric Oxide Synthase Type II/metabolism , Peptides/pharmacology , Venoms/pharmacology , 3' Untranslated Regions , Animals , Cell Line , Dactinomycin/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Enzyme Stability , Exenatide , Gene Expression Regulation, Enzymologic , Glucagon-Like Peptide-1 Receptor , Imines/pharmacology , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitrites/metabolism , Promoter Regions, Genetic , Protein Binding , Proteolysis , RNA Stability , Receptors, Glucagon/agonists
5.
Biochem Biophys Res Commun ; 439(2): 258-63, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23973482

ABSTRACT

The activation of pancreatic stellate cells (PSCs) is thought to be a potential mechanism underlying islet fibrosis, which may contribute to progressive ß-cell failure in type 2 diabetes. Recently, we demonstrated that antioxidants reduced islet fibrosis in an animal model of type 2 diabetes. However, there is no in vitro study demonstrating that high glucose itself can induce oxidative stress in PSCs. Thus, PSCs were isolated and cultured from Sprague Dawley rats, and treated with high glucose for 72 h. High glucose increased the production of reactive oxygen species. When treated with high glucose, freshly isolated PSCs exhibited myofibroblastic transformation. During early culture (passage 1), PSCs treated with high glucose contained an increased number of α-smooth muscle actin-positive cells. During late culture (passages 2-5), PSCs treated with high glucose exhibited increases in cell proliferation, the expression of fibronectin and connective tissue growth factor, release of interleukin-6, transforming growth factor-ß and collagen, and cell migration. Finally, the treatment of PSCs with high glucose and antioxidants attenuated these changes. In conclusion, we demonstrated that high glucose increased oxidative stress in primary rat PSCs, thereby facilitating the activation of these cells, while antioxidant treatment attenuated high glucose-induced PSC activation.


Subject(s)
Glucose/metabolism , Oxidative Stress , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Animals , Antioxidants/pharmacology , Cell Movement/drug effects , Cells, Cultured , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Fibrosis/metabolism , Fibrosis/pathology , Male , Oxidative Stress/drug effects , Pancreas/cytology , Pancreas/metabolism , Pancreas/pathology , Pancreatic Stellate Cells/cytology , Pancreatic Stellate Cells/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
6.
Mol Cell Endocrinol ; 572: 111947, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37150285

ABSTRACT

Hypoxia in pancreatic islets (islet hypoxia) can occur in type 2 diabetes mellitus. Previously, our in vitro experiments demonstrated that pancreatic stellate cells (PSCs) within the islet are activated in hypoxia, promoting pancreatic ß-cell death. Here, we aimed to demonstrate the in vivo activation of intra-islet PSCs and investigate the mechanism of PSC-induced ß-cell death in hypoxia. A novel in vivo model of islet hypoxia was established by injecting fluorescent microspheres into a carotid artery of Balb/c mice (Microsphere mice). The intraperitoneal glucose tolerance (IPGTT) was performed, and pancreatic tissues were stained for insulin expression after tissue clearing. Pimonidazole staining was also performed in the pancreas to detect the presence of hypoxia in islets. Next, primary PSCs were isolated and cultured from Balb/c mice. Exosomes were isolated from culture media from PSCs cultured in hypoxia (1% oxygen). MicroRNAs (miRNAs) were prepared from exosomes from PSCs, and miRNA expression profiles were analyzed by miRNA sequencing. Several miRNAs were overexpressed in islets using miRNA mimics. Two weeks after injection of microspheres, the Microsphere mice showed worsening of glucose tolerance in IPGTT. Later, cataracts were developed in the eyes of the mice. The pancreas showed that the areas, perimeters, and diameters of insulin-positive cells decreased in Microsphere mice. Pimonidazole adducts were detected in the islets of these mice, indicating the presence of islet hypoxia. In addition, α-smooth muscle actin-positive cell numbers per islet were higher in Microsphere mice, confirming the in vivo activation of intra-islet PSCs in hypoxia. Mouse islets incubated with exosomes isolated from PSCs cultured in hypoxia showed a decrease in cell viability. The exosomes contained a variety of miRNAs, of which miR-23a-3p was found to notably increase ß-cell death through apoptosis. Together, our in vivo and in vitro data provide evidence to support that PSCs within the islets are activated in hypoxia and promote ß-cell death through exosomal miRNA transfer, which may contribute to the progression of type 2 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Type 2 , Islets of Langerhans , MicroRNAs , Mice , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Diabetes Mellitus, Type 2/metabolism , Pancreatic Stellate Cells/metabolism , Islets of Langerhans/metabolism , Insulin/metabolism , Glucose/metabolism , Hypoxia/metabolism , Cell Death
7.
Plant Physiol Biochem ; 200: 107804, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37269823

ABSTRACT

The tomato (Solanum lycopersicum) is widely consumed globally and renowned for its health benefits, including the reduction of cardiovascular disease and prostate cancer risk. However, tomato production faces significant challenges, particularly due to various biotic stresses such as fungi, bacteria, and viruses. To address this challenges, we employed the CRISPR/Cas9 system to modify the tomato NUCLEOREDOXIN (SlNRX) genes (SlNRX1 and SlNRX2) belonging to the nucleocytoplasmic THIOREDOXIN subfamily. CRISPR/Cas9-mediated mutations in SlNRX1 (slnrx1) plants exhibited resistance against bacterial leaf pathogen Pseudomonas syringae pv. maculicola (Psm) ES4326, as well as the fungal pathogen Alternaria brassicicola. However, the slnrx2 plants did not display resistance. Notably, the slnrx1 demonstrated elevated levels of endogenous salicylic acid (SA) and reduced levels of jasmonic acid after Psm infection, in comparison to both wild-type (WT) and slnrx2 plants. Furthermore, transcriptional analysis revealed that genes involved in SA biosynthesis, such as ISOCHORISMATE SYNTHASE 1 (SlICS1) and ENHANCED DISEASE SUSCEPTIBILITY 5 (SlEDS5), were upregulated in slnrx1 compared to WT plants. In addition, a key regulator of systemic acquired resistance, PATHOGENESIS-RELATED 1 (PR1), exhibited increased expression in slnrx1 compared to WT. These findings suggest that SlNRX1 acts as a negative regulator of plant immunity, facilitating infection by the Psm pathogen through interference with the phytohormone SA signaling pathway. Thus, targeted mutagenesis of SlNRX1 is a promising genetic means to enhance biotic stress resistance in crop breeding.


Subject(s)
Salicylic Acid , Solanum lycopersicum , Salicylic Acid/metabolism , Solanum lycopersicum/genetics , Plant Breeding , Pseudomonas syringae/physiology , Signal Transduction/genetics , Cyclopentanes/metabolism , Plant Diseases/microbiology , Gene Expression Regulation, Plant
8.
Microorganisms ; 10(11)2022 Nov 14.
Article in English | MEDLINE | ID: mdl-36422322

ABSTRACT

The emergence of antibiotic resistance in foodborne pathogens isolated from meat pro-ducts and their producing environment has been an increasing and leading threat to public health. The aim of the study was to identify pathogens and their antimicrobial resistance isolated from pig production to pork meat distribution phases. Through this study, food spoilage and foodborne or clinical pathogenic bacteria were isolated and identified from pork (belly and neck) meat product and its related environmental samples that include pig swabs, diets, feces, liquid manure, workers' gloves, dust fan swabs, carcass swabs, floor swabs, and drain water in the affiliated farm, slaughterhouse, meat processing plant, and in retail stores. All carcasses at the slaughterhouse and meat products at the meat processing plant were tracked from pigs at a targeted farm. Nine different selective media agars were used to effectively isolate various pathogenic bacteria. A total of 283 presumptive pathogenic bacteria isolated from 126 samples were selected and identified using MALDI-ToF MS. Twenty-three important foodborne pathogens were identified, and some of them, Shiga-toxin-producing E. coli (STEC), Listeria monocytogenes, Staphylococcus aureus, and Yersinia enterocolitica, were further confirmed using PCR. The PFGE patterns of 12 STEC isolates were grouped by sample source or site. All the foodborne pathogens used in the study were not resistant to amoxicillin/clavulanate, ciprofloxacin, and gentamicin, whereas some of the STEC, L. monocytogenes, and S. aureus isolates were resistant to various antibiotics, including ampicillin, erythromycin, tetracycline, and vancomycin. The most common antimicrobial resistance pattern in the pathogenic STEC isolates was AMP-KAN-STR-SXT-TET. Consequently, this study provides valuable information for the distribution of antimicrobial-resistant pathogens along the pork meat production chain and can assist farmers and stakeholders to develop a systematic strategy for reducing the current emergence and spread of antimicrobial resistance in the different phases of pig production and distribution.

9.
Biochem Biophys Res Commun ; 414(2): 397-402, 2011 Oct 22.
Article in English | MEDLINE | ID: mdl-21971557

ABSTRACT

Islet fibrosis could be important in the progression of pancreatic beta cell failure in type 2 diabetes. It is known that oxidative stress is involved in the pancreatic fibrosis through the activation of pancreatic stellate cells. However, no study has investigated the in vivo effects of antioxidants on islet fibrogenesis in type 2 diabetes. In this study, antioxidants (taurine or tempol) were administered in drinking water to Otsuka Long-Evans Tokushima Fatty rats, an animal model of type 2 diabetes, for 16 weeks. An intraperitoneal glucose tolerance test revealed that the blood glucose levels after the glucose injection were decreased by the antioxidants. The insulin secretion after the glucose injection, which was markedly reduced in the rats, was also restored by the antioxidants. Beta cell mass and pancreatic insulin content were greater in the rats treated with the antioxidants than in the untreated rats. Beta cell apoptosis was attenuated in the rats by the antioxidants. Finally, islet fibrosis and the activation of pancreatic stellate cells were markedly diminished in the rats by the antioxidants. Our data suggest that antioxidants may protect beta cells through the attenuation of both islet fibrosis and beta cell apoptosis in type 2 diabetes.


Subject(s)
Antioxidants/administration & dosage , Apoptosis/drug effects , Cytoprotection , Diabetes Mellitus, Experimental/prevention & control , Diabetes Mellitus, Type 2/prevention & control , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Animals , Blood Glucose/analysis , Cyclic N-Oxides/administration & dosage , Fibrosis , Glucose Tolerance Test , Insulin/metabolism , Insulin Secretion , Male , Rats , Rats, Inbred OLETF , Spin Labels , Taurine/administration & dosage
10.
Cell Physiol Biochem ; 25(2-3): 211-20, 2010.
Article in English | MEDLINE | ID: mdl-20110682

ABSTRACT

High doses of glucosamine have been known to induce apoptosis of pancreatic beta cells. The mechanism for this phenomenon has not been clearly elucidated. We aimed to explore the potential mechanisms for glucosamine toxicity in the rat insulinoma cell line INS-1 and in rat native beta cells. We also investigated whether glucagon-like peptide (GLP)-1 could be protective against glucosamine. Glucosamine exhibited dose-dependent inhibition of cell survival and an increase in the cell population at the sub-G1 phase. Glucosamine was revealed to inhibit cellular glucose uptake, resulting in the activation of AMP-activated protein kinase (AMPK). Accordingly, phosphorylation of P70S6K and ribosomal protein S6 (S6RP) was decreased. Protein glycosylation appeared not to be involved in this cytotoxicity. Pretreatment with GLP-1 alleviated glucosamine-mediated inhibition of glucose uptake and lessened AMPK activation, thus allowing recovery of the phosphorylation levels of P70S6K and S6RP. The effect of GLP-1 was blocked by the adenylyl cyclase inhibitor MDL12330A but not by the protein kinase A inhibitor H89. Taken together, these data demonstrate that glucosamine may inhibit beta-cell survival by diminishing cellular glucose uptake independent of glycosylation. This glucosamine toxicity can be blocked by GLP-1, which leads to recovery of the glucose uptake through a PKA-independent, cAMP-dependent mechanism.


Subject(s)
Glucagon-Like Peptide 1/physiology , Glucosamine/toxicity , Insulin-Secreting Cells/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis , Cell Line, Tumor , G1 Phase , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Phosphorylation , Rats , Ribosomal Protein S6/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
11.
Diabetes Metab J ; 44(6): 919-927, 2020 12.
Article in English | MEDLINE | ID: mdl-32431113

ABSTRACT

BACKGROUND: Hypoxia can occur in pancreatic islets in type 2 diabetes mellitus. Pancreatic stellate cells (PSCs) are activated during hypoxia. Here we aimed to investigate whether PSCs within the islet are also activated in hypoxia, causing ß-cell injury. METHODS: Islet and primary PSCs were isolated from Sprague Dawley rats, and cultured in normoxia (21% O2) or hypoxia (1% O2). The expression of α-smooth muscle actin (α-SMA), as measured by immunostaining and Western blotting, was used as a marker of PSC activation. Conditioned media (hypoxia-CM) were obtained from PSCs cultured in hypoxia. RESULTS: Islets and PSCs cultured in hypoxia exhibited higher expressions of α-SMA than did those cultured in normoxia. Hypoxia increased the production of reactive oxygen species. The addition of N-acetyl-L-cysteine, an antioxidant, attenuated the hypoxia-induced PSC activation in islets and PSCs. Islets cultured in hypoxia-CM showed a decrease in cell viability and an increase in apoptosis. CONCLUSION: PSCs within the islet are activated in hypoxia through oxidative stress and promote islet cell death, suggesting that hypoxia-induced PSC activation may contribute to ß-cell loss in type 2 diabetes mellitus.


Subject(s)
Pancreatic Stellate Cells , Animals , Apoptosis , Cell Hypoxia , Cells, Cultured , Diabetes Mellitus, Type 2 , Hypoxia , Rats , Rats, Sprague-Dawley
12.
Plant Physiol Biochem ; 156: 105-114, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32927152

ABSTRACT

Many stresses induce the accumulation of unfolded and misfolded proteins in the endoplasmic reticulum, a phenomenon known as ER stress. In response to ER stress, cells initiate a protective response, known as unfolded protein response (UPR), to maintain cellular homeostasis. The UPR sensor, inositol-requiring enzyme 1 (IRE1), catalyzes the cytoplasmic splicing of bZIP transcription factor-encoding mRNAs to activate the UPR signaling pathway. Recently, we reported that pretreatment of Arabidopsis thaliana plants with tunicamycin, an ER stress inducer, increased their susceptibility to bacterial pathogens; on the other hand, IRE1 deficient mutants were susceptible to Pseudomonas syringae pv. maculicola (Psm) and failed to induce salicylic acid (SA)-mediated systemic acquired resistance. However, the functional relationship of IRE1 with the pathogen and TM treatment remains unknown. In the present study, we showed that bacterial pathogen-associated molecular patterns (PAMPs) induced IRE1 expression; however, PAMP-triggered immunity (PTI) response such as callose deposition, PR1 protein accumulation, or Pst DC3000 hrcC growth was not altered in ire1 mutants. We observed that IRE1 enhanced plant immunity against the bacterial pathogen P. syringae pv. tomato DC3000 (Pst DC3000) under ER stress. Moreover, TM-pretreated ire1 mutants were more susceptible to the avirulent strain Pst DC3000 (AvrRpt2) and showed greater cell death than wild-type plants during effector-triggered immunity (ETI). Additionally, Pst DC3000 (AvrRpt2)-mediated RIN4 degradation was reduced in ire1 mutants under TM-induced ER stress. Collectively, our results reveal that IRE1 plays a pivotal role in the immune signaling pathway to activate plant immunity against virulent and avirulent bacterial strains under ER stress.


Subject(s)
Arabidopsis Proteins/immunology , Arabidopsis/enzymology , Endoplasmic Reticulum Stress , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/immunology , Arabidopsis/immunology , Arabidopsis Proteins/metabolism , Bacterial Proteins , Inositol , Pathogen-Associated Molecular Pattern Molecules/metabolism , Plant Diseases/immunology , Plant Diseases/microbiology , Plant Immunity , Pseudomonas syringae , Signal Transduction
13.
Biochem Biophys Res Commun ; 386(2): 356-62, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19523921

ABSTRACT

In islet transplantation, a substantial part of the graft becomes nonfunctional for several reasons including hypoxia. AMP-activated protein kinase (AMPK) in mammalian cells is a regulator of energy homeostasis, and is activated by metabolic stresses such as hypoxia. However, the role of AMPK in hypoxic injury to pancreatic beta cells is not clear. When a rat beta cell line, INS-1 cell, was incubated in an anoxic chamber, phosphorylation of both AMPK and its downstream protein, acetyl-CoA carboxylase 2 increased with time. Adenovirus-mediated expression of constitutively active form of AMPK under normoxic conditions increased caspase-3 activation, suggesting induction of apoptosis. Reactive oxygen species production also increased with time during hypoxia. Pretreatment with compound C, an AMPK inhibitor, or N-acetyl-l-cysteine, an antioxidant, significantly lowered hypoxia-mediated cell death. These results suggest that AMPK, in association with oxidative stress, plays an important role in acute and severe hypoxic injury to pancreatic beta cells.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Hypoxia/enzymology , Insulin-Secreting Cells/enzymology , Animals , Apoptosis , Cell Line , Enzyme Activation , Hypoxia/pathology , Insulin-Secreting Cells/pathology , Rats , Reactive Oxygen Species/metabolism
14.
J Cell Biochem ; 104(6): 2261-71, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18446785

ABSTRACT

Glucagon-like peptide-1 (GLP-1) induces several immediate early response genes such as c-fos, c-jun, and early growth response-1 (Egr-1), which are involved in cell proliferation and differentiation. We recently reported that exendin-4 (EX-4), a potent GLP-1 agonist, upregulated Egr-1 expression via phosphorylation of CREB, a transcription factor in INS-1 beta-cells. This study was designed to investigate the role of another transcription factors, serum response factor (SRF) and Yin Yang-1 (YY1), in EX-4-induced Egr-1 expression. EX-4 significantly increased Egr-1 mRNA and subsequently its protein level. EX-4-induced Egr-1 expression was inhibited by pretreatment with a PKA inhibitor, H-89, and an MEK inhibitor, PD 98059. The siRNA-mediated inhibition of PKA and ERK1 resulted in significant reduction of EX-4-induced Egr-1 expression. Promoter analyses showed that SRE clusters were essential for Egr-1 transcription, and YY1 overexpression did not affect Egr-1 promoter activity. EMSA results demonstrated that EX-4-induced transient increase in DNA-protein complex on SRE site, and that both SRF and phospho-SRF were bound to this site. Treatment of either YY1 consensus oligonucleotide or YY1 antibody did not effect the change of density or migration of the DNA-protein complex. Collectively, EX-4-induced Egr-1 expression is largely dependent on cAMP-mediated extracellular signal-regulated kinase activation, and EX-4 induces Egr-1 transcription via the interaction of SRF and phospho-SRF to SRE sites.


Subject(s)
Early Growth Response Protein 1/genetics , Gene Expression Regulation/drug effects , Insulin-Secreting Cells/metabolism , Peptides/pharmacology , Response Elements/genetics , Serum Response Factor/metabolism , Venoms/pharmacology , YY1 Transcription Factor/metabolism , Animals , Cell Line , Cyclic AMP/metabolism , Early Growth Response Protein 1/metabolism , Electrophoretic Mobility Shift Assay , Exenatide , Extracellular Signal-Regulated MAP Kinases/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/enzymology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Transcription, Genetic/drug effects
15.
Transplantation ; 85(3): 323-30, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18301327

ABSTRACT

BACKGROUND: Islet transplantation is a potential strategy to cure type 1 diabetes mellitus. However, a substantial part of the islet graft becomes nonfunctional due to several factors including hypoxia. However, the precise mechanism of cell damage is largely unknown in hypoxic exposure to pancreatic beta cells. The aim of the present study was to investigate whether acute and severe hypoxic injury could involve inducible nitric oxide synthase (iNOS)-nitric oxide (NO) signaling in beta cells. METHODS: The rat beta cell line (INS-1) and primary rat islets were incubated in an anoxic chamber. Cell viability was determined by propium iodide staining or cell counting kit. The expression of iNOS mRNA and protein was examined using reverse-transcription polymerase chain reaction and Western blot analysis. NO production was measured as nitrite accumulation by Griess reagent method. RESULTS: After hypoxic exposure, marked cell death occurred in INS-1 cells and rat islets, accompanied by increase in activated caspase-3 expression. NO production was increased in the culture medium in a time-dependent manner. Increase in expression of iNOS mRNA and protein was found. Pretreatment with a selective iNOS inhibitor, 1400W, significantly prevented cell death during hypoxia. In addition, hypoxia activated c-Jun N-terminal kinase (JNK) significantly, but the addition of 1400W inhibited hypoxia-induced JNK phosphorylation. CONCLUSIONS: Our data suggest that iNOS-NO plays an important role in acute and severe hypoxic injury to pancreatic beta cells. Therefore, iNOS-NO might be a potential therapeutic target for preserving beta cell survival in islet transplantation through prevention of hypoxia-mediated cell death.


Subject(s)
Insulin-Secreting Cells/enzymology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/biosynthesis , Animals , Caspase 3/metabolism , Cell Hypoxia , Cell Separation , Cell Survival , Cells, Cultured , Enzyme Activation , Gene Expression Regulation, Enzymologic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin-Secreting Cells/cytology , Male , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide Synthase Type II/genetics , Rats , Rats, Sprague-Dawley
16.
PLoS One ; 13(10): e0206401, 2018.
Article in English | MEDLINE | ID: mdl-30379923

ABSTRACT

Various subtypes of enteroendocrine cells (EECs) are present in the gut epithelium. EECs and pancreatic ß-cells share similar pathways of differentiation during embryonic development and after birth. In this study, similarities between EECs and ß-cells were evaluated in detail. To obtain specific subtypes of EECs, cell sorting by flow cytometry was conducted from STC-1 cells (a heterogenous EEC line), and each single cell was cultured and passaged. Five EEC subtypes were established according to hormone expression, measured by quantitative RT-PCR and immunostaining: L, K, I, G and S cells expressing glucagon-like peptide-1, glucose-dependent insulinotropic polypeptide, cholecystokinin, gastrin and secretin, respectively. Each EEC subtype was found to express not only the corresponding gut hormone but also other gut hormones. Global microarray gene expression profiles revealed a higher similarity between each EEC subtype and MIN6 cells (a ß-cell line) than between C2C12 cells (a myoblast cell line) and MIN6 cells, and all EEC subtypes were highly similar to each other. Genes for insulin secretion-related proteins were mostly enriched in EECs. However, gene expression of transcription factors crucial in mature ß-cells, such as PDX1, MAFA and NKX6.1, were remarkably low in all EEC subtypes. Each EEC subtype showed variable methylation in three cytosine-guanosine dinucleotide sites in the insulin gene (Ins2) promoter, which were fully unmethylated in MIN6 cells. In conclusion, our data confirm that five EEC subtypes are closely related to ß-cells, suggesting a potential target for cell-based therapy in type 1 diabetes.


Subject(s)
DNA Methylation , Enteroendocrine Cells/metabolism , Gene Expression Profiling , Insulin-Secreting Cells/metabolism , Insulin/genetics , Animals , Cell Line , Mice , Transcription Factors/genetics
17.
Biochem Pharmacol ; 68(9): 1775-85, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15450943

ABSTRACT

Cytokines that are released by infiltrating inflammatory cells around the pancreatic islets are involved in the pathogenesis of type 1 diabetes mellitus. Specifically, interleukin-1beta (IL-1beta) stimulates inducible nitric oxide synthase (iNOS) expression and nitric oxide overproduction, leading to the beta-cell damage. In activating this pathway, nuclear factor-kappaB (NF-kappaB) plays a crucial role, and many of the IL-1beta-sensitive genes contain NF-kappaB binding sites in their promoter regions. We have recently shown that epicatechin, which is a flavonoid, had a protective effect on pancreatic beta-cells in both streptozotocin-treated rats and islets. In the present study, the effects of epicatechin on IL-1beta-induced beta-cell damage were examined. RINm5F cells and islets were pretreated with epicatechin and next incubated with IL-1beta. The released nitrite, iNOS protein and mRNA expression levels were then measured. IkappaBalpha protein, nuclear translocation of NF-kappaB, and NF-kappaB DNA binding activity were also determined. Following the transient transfection of an iNOS promoter into the cells, the iNOS promoter activity was measured. ATP- or D-glucose-induced insulin release was measured in RINm5F cells and islets, respectively. Epicatechin significantly reduced IL-1beta-induced nitrite production, iNOS protein and mRNA expressions, and it also inhibited IL-1beta-induced IkappaBalpha protein degradation, NF-kappaB activation, and iNOS promoter activity. Epicatechin partly restored the IL-1beta-induced inhibition of insulin release. These results suggest that epicatechin inhibits the IL-1beta-induced iNOS expression by down-regulating NF-kappaB activation, and protecting beta-cells from IL-1beta.


Subject(s)
Catechin/pharmacology , Interleukin-1/pharmacology , Islets of Langerhans/drug effects , NF-kappa B/metabolism , Nitric Oxide Synthase/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Down-Regulation , Gene Expression/drug effects , I-kappa B Proteins/metabolism , I-kappa B Proteins/pharmacology , Insulin/metabolism , Islets of Langerhans/enzymology , NF-KappaB Inhibitor alpha , Nitric Oxide/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats
18.
Korean J Gastroenterol ; 42(6): 519-26, 2003 Dec.
Article in Korean | MEDLINE | ID: mdl-14695709

ABSTRACT

BACKGROUND/AIMS: Pancreatic acini of streptozotocin (STZ)-induced diabetic rats release amylase less than normal acini on cholecystokinin (CCK) stimulation. Pancreatic enzyme secretion has been closely related to the intracellular calcium concentration ([Ca2(+)](i)) of the acinar cell. In the present study, sequential changes of the intracellular calcium signal which probably underlie the altered enzyme secretion in response to CCK-8 were investigated using pancreatic acini from diabetic rats. METHODS: Diabetic rats were prepared by single intravenous injection of STZ (70 mg/kg). Stimulating experiments with CCK-8 were performed 7 days later. Pancreatic acini were isolated by collagenase digestion. Amylase release and [Ca2(+)](i) were measured by colorimethod and calcium imaging, respectively. The geometry of intracellular calcium signal was analyzed. RESULTS: Normal acini exhibited concentration-dependent [Ca2(+)](i) increase and regular oscillatory calcium signal on CCK-8 stimulation. Amylase release was also concentration-dependent. However, diabetic acini showed significantly less [Ca2(+)](i) increase, prolonged time to peak [Ca2(+)](i), decreased calcium spikes number, and decreased amylase release compared with normal acini. The decreased [Ca2(+)](i) in diabetic acini was restored significantly by insulin treatment. CONCLUSIONS: Relatively decreased amylase release in diabetic pancreatic acini in response to CCK, appears to be associated with altered calcium signal due to insulin deficiency.


Subject(s)
Amylases/metabolism , Calcium Signaling/drug effects , Diabetes Mellitus, Experimental/physiopathology , Pancreas/metabolism , Sincalide/pharmacology , Animals , Pancreas/cytology , Rats , Rats, Sprague-Dawley
19.
Diabetes Metab J ; 35(1): 65-71, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21537415

ABSTRACT

BACKGROUND: Type 2 diabetes mellitus (T2DM) is often accompanied by increased levels of circulating fatty acid. Elevations in fatty acids and glucose for prolonged periods of time have been suggested to cause progressive dysfunction or apoptosis of pancreatic beta cells in T2DM. However, the precise mechanism of this adverse effect is not well understood. METHODS: INS-1 rat-derived insulin-secreting cells were exposed to 30 mM glucose and 0.25 mM palmitate for 48 hours. RESULTS: The production of reactive oxygen species increased significantly. Pancreatic and duodenal homeobox 1 (Pdx1) expression was down-regulated, as assessed by reverse transcription-polymerase chain reaction and Western blot analyses. The promoter activities of insulin and Pdx1 were also diminished. Of note, there was nucleocytoplasmic translocation of Pdx1, which was partially prevented by treatment with an antioxidant, N-acetyl-L-cysteine. CONCLUSION: Our data suggest that prolonged exposure of beta cells to elevated levels of glucose and palmitate negatively affects Pdx1 expression via oxidative stress.

20.
Diabetes Metab J ; 35(5): 469-79, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22111038

ABSTRACT

BACKGROUND: Glucose toxicity that is caused by chronic exposure to a high glucose concentration leads to islet dysfunction and induces apoptosis in pancreatic ß-cells. Heme oxygenase-1 (HO-1) has been identified as an anti-apoptotic and cytoprotective gene. The purpose of this study is to investigate whether HO-1 up-regulation when using metalloprotophyrin (cobalt protoporphyrin, CoPP) could protect pancreatic ß-cells from high glucose-induced apoptosis. METHODS: Reverse transcription-polymerase chain reaction was performed to analyze the CoPP-induced mRNA expression of HO-1. Cell viability of INS-1 cells cultured in the presence of CoPP was examined by acridine orange/propidium iodide staining. The generation of intracellular reactive oxygen species (ROS) was measured using flow cytometry. Glucose stimulated insulin secretion (GSIS) was determined following incubation with CoPP in different glucose concentrations. RESULTS: CoPP increased HO-1 mRNA expression in both a dose- and time-dependent manner. Overexpression of HO-1 inhibited caspase-3, and the number of dead cells in the presence of CoPP was significantly decreased when exposed to high glucose conditions (HG). CoPP also decreased the generation of intracellular ROS by 50% during 72 hours of culture with HG. However, decreased GSIS was not recovered even in the presence of CoPP. CONCLUSION: Our data suggest that CoPP-induced HO-1 up-regulation results in protection from high glucose-induced apoptosis in INS-1 cells; however, glucose stimulated insulin secretion is not restored.

SELECTION OF CITATIONS
SEARCH DETAIL