Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Pharmacol Rep ; 70(4): 677-683, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29940507

ABSTRACT

BACKGROUND: Many studies have indicated a relationship between diabetes and Alzheimer's disease (AD). However, the molecular mechanism underlying this association has not been clarified. Among several factors, insulin degrading enzyme (IDE), which plays roles in the degradation of both insulin and amyloid ß (Aß), has gained interest as a potential target in efforts to solve this puzzle. This study sought to examine the effects of varying insulin and/or glucose concentrations on IDE expression. METHODS: Experiments were performed on primary cultured rat neurons and cortices of rats with streptozotocin (STZ)-induced diabetes. IDE protein and mRNA expression levels were measured by western blot and RT-PCR, respectively. RESULTS: In primary cultured cortical neurons, removal of insulin for 5days reduced the expression of IDE. A five-day treatment with a high concentration of glucose in insulin-free media reduced IDE levels, while a high concentration of glucose in the presence of insulin had no effect. In groups treated with glucose or insulin intermittently, the reduction in IDE levels was observed only in neurons exposed to high glucose together with no insulin for 5days. Shorter incubation periods (48h), either continuously or intermittently, did not affect IDE levels. IDE expression in the cortex of rats with STZ-induced diabetes was found to be decreased. CONCLUSION: Our data suggest that insulin deprivation, rather than high glucose, is a significant determinant of IDE regulation. As evidence indicates potential roles for IDE in diabetes and AD, understanding the mechanisms regulating IDE expression may be important in developing new treatment strategies.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Diabetes Mellitus, Experimental/metabolism , Insulin/deficiency , Insulysin/biosynthesis , Neurons/metabolism , Animals , Cerebral Cortex/enzymology , Diabetes Mellitus, Experimental/enzymology , Dose-Response Relationship, Drug , Glucose/pharmacology , Insulin/metabolism , Male , Neurons/enzymology , Primary Cell Culture , Rats , Time Factors
2.
Histol Histopathol ; 33(11): 1167-1180, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29726577

ABSTRACT

Insulin Degrading Enzyme (IDE) is an endopeptidase that degrades insulin and glucagon. Ide gene has been associated with type-2 diabetes mellitus (DM2). However, the physiological role(s) of IDE in glucose homeostasis and its potential therapeutic benefit remain not completely known. To contribute in the understanding of IDE's role in glucose metabolism, we analyzed IDE protein level in pancreatic islets from two hyperinsulinemic mouse models, db/db and high-fat diet (HFD) mice, as well as in human islets from DM2 patients treated with oral hypoglycemic agents (OHAs) or insulin. IDE protein level was detected by staining and by western-blot. INS1E cells, rat and human islets were treated with insulin and IDE protein level was studied. We have shown for the first time IDE staining in rodent and human tissue, using the proper negative control, IDE null mouse tissue. Our staining indicates that IDE is expressed in both beta- and alpha-cells, with higher expression in alpha-cells. Db/db and HFD mice islets showed increased IDE protein level. Interestingly, human islets from DM2 patients treated with OHAs showed decreased IDE protein level in beta-cells. Meanwhile, islets from insulin-treated DM2 patients showed augmented IDE protein level compared to OHAs patients, pointing to an upregulation of IDE protein level stimulated by insulin. These data correlate nicely with insulin-stimulated upregulation of IDE in cultured INS1E cells, as well as in rat and human islets. In conclusion, our study shows that IDE is expressed in pancreatic beta- and alpha-cells of both rodents and humans, having higher expression in alpha-cells. Furthermore, insulin stimulates IDE protein level in pancreatic beta-cells. These results may have implications in how DM2 patient's treatment affects their beta-cell function.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucagon-Secreting Cells/enzymology , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/enzymology , Insulin/pharmacology , Insulysin/biosynthesis , Islets of Langerhans/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Humans , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Mice , Up-Regulation
3.
Prep Biochem Biotechnol ; 45(6): 596-604, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-25036570

ABSTRACT

Inulinase production by Rhodotorula glutinis was carried out in this study, using leek (Allium ampeloprasum var. porrum) as an alternative carbon source due to its high inulin content and easy availability. Taguchi orthogonal array (OA) design of experiment (DOE) was used to optimize fermentation conditions. For this purpose, five influential factors (leek concentration, pH, incubation temperature, agitation speed, and fermentation time) related to inulinase production were selected at four convenient levels. The results showed that maximum inulinase activity was obtained as 30.89 U/mL, which was close to the predicted result (30.24 U/mL). To validate the obtained results, analysis of variance (ANOVA) was employed. Consequently, leek has a great potential as an effective and economical carbon source for inulinase production, and the use of Taguchi DOE enhanced enzyme activity about 2.87-fold when compared with the unoptimized condition.


Subject(s)
Allium/microbiology , Bioreactors/microbiology , Insulysin/biosynthesis , Insulysin/isolation & purification , Rhodotorula/enzymology , Rhodotorula/growth & development
4.
Carbohydr Polym ; 102: 662-8, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24507333

ABSTRACT

In this study, screening and optimization of nutrients for inulinase production using copra waste has been studied. Plackett-Burman Design (PBD) was employed to screen the significant nutrients for inulinase production. Response surface methodology (RSM) was used to evaluate the effects of nutrient components in the medium. The second order regression equation provides the inulinase activity as the function of K2HPO4, ZnSO4 · 7H2O and soya bean cake. The optimum conditions are: K2HPO4--0.0047 g/gds, ZnSO4 · 7H2O - 0.02677 g/gds and soya bean cake--0.06288 g/gds. At these optimized conditions, experiments were performed in packed bed bioreactor to optimize the process variables like air flow rate, packing density, particle size and moisture content. The optimum conditions were: air flow rate--0.76 L/min, packing density--38 g/L, particle size--10/14 mesh and moisture content--60%. At the optimized conditions, a maximum inulinase production of 239 U/gds was achieved.


Subject(s)
Bioreactors , Fermentation , Industrial Waste , Insulysin/biosynthesis , Fungi/metabolism
5.
J Cereb Blood Flow Metab ; 34(2): 258-67, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24301291

ABSTRACT

To better understand the role of downstream Toll-like receptor (TLR) signaling during acute cerebral ischemia, we performed cDNA microarrays, on brain RNA, and cytokine arrays, on serum, from wild type (WT), MyD88-/- and TRIF-mutant mice, at baseline and following permanent middle cerebral artery occlusion (pMCAO). The acute stress response pathway was among the top pathways identified by Ingenuity Pathway Analysis of microarray data. We used real-time polymerase chain reaction to confirm the expression of four immediate early genes; EGR1, EGR2, ARC, Nurr77, in this pathway, and insulin degrading enzyme (IDE). Compared to WT, baseline immediate early gene expression was increased up to10-fold in MyD88-/- and TRIF-mutant mice. However, following pMCAO, immediate early gene expression remained unchanged, from this elevated baseline in these mice, but increased up to 12-fold in WT. Furthermore, expression of IDE, which also degrades ß-amyloid, decreased significantly only in TRIF-mutant mice. Finally, sE-Selectin, sICAM, sVCAM-1, and MMP-9 levels were significantly decreased only in MyD88-/- compared with WT mice. We thus report a new role for downstream TLR signaling in immediate early gene expression during acute cerebral ischemia. We also show that the TRIF pathway regulates IDE expression; a major enzyme that clears ß-amyloid from the brain.


Subject(s)
Signal Transduction , Toll-Like Receptors/metabolism , Acute Disease , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/metabolism , Gene Expression Regulation, Enzymologic/genetics , Insulysin/biosynthesis , Insulysin/genetics , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Toll-Like Receptors/genetics
6.
Acta Anaesthesiol Scand ; 57(7): 903-10, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23495897

ABSTRACT

BACKGROUND: Patients with Alzheimer's disease (AD) exhibit a failure in the clearance of amyloid ß peptides (Aß) from the central nervous system. Previous studies have suggested an association between anesthesia and the occurrence of AD. The aim of the present report was to further explore this possibility. METHODS: Animals were administered sevoflurane for 2 h. We performed immunohistochemistry and real-time polymerase chain reaction to assess the levels of low-density lipoprotein receptor-related protein 1 (LRP-1), the receptor for advanced glycation end products (RAGE) protein, insulin-degrading enzyme (IDE), and neprilysin (NEP) in aged and young rat's brain. RESULT: Levels of LRP-1 were significantly decreased, while those of RAGE increased in the aged and young groups. Immunoreactivity for IDE was significantly decreased at 3 and increased at 15 days in the young group. In contrast, immunoreactivity for NEP was significantly increased at 1 but decreased at 15 days in aged rats. Levels of IDE messenger RNA (mRNA) were significantly decreased at 3 and 7 days in the aged group but was consistently decreased at 1, 3, 7, and 15 days in the young group. Levels of NEP mRNA were significantly decreased in the aged group but increased in the young group at 1, 3, 7, and 15 days. CONCLUSION: Sevoflurane leads to a reduction in the levels of LRP-1, while increasing RAGE and decreasing IDE and NEP in both aged and, to a lesser extent, young rat's brain. These receptor and enzymatic changes may promote the accumulation of Aß in brain tissues and thus exacerbate Alzheimer's-like pathology.


Subject(s)
Amyloid beta-Peptides/metabolism , Anesthetics, Inhalation/toxicity , Brain Chemistry/drug effects , Methyl Ethers/toxicity , Nerve Tissue Proteins/analysis , Aging/metabolism , Alzheimer Disease/metabolism , Animals , Blood-Brain Barrier/drug effects , Brain/enzymology , CA1 Region, Hippocampal/chemistry , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/enzymology , Female , Insulysin/analysis , Insulysin/biosynthesis , Insulysin/genetics , Low Density Lipoprotein Receptor-Related Protein-1/analysis , Low Density Lipoprotein Receptor-Related Protein-1/biosynthesis , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Neprilysin/analysis , Neprilysin/biosynthesis , Neprilysin/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptor for Advanced Glycation End Products , Receptors, Immunologic/analysis , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , Sevoflurane , Time Factors
7.
PLoS One ; 8(3): e59155, 2013.
Article in English | MEDLINE | ID: mdl-23520555

ABSTRACT

BACKGROUND AND PURPOSE: The present study was designed to examine the effects of ginsenoside Rg1 on expression of peroxisome proliferator-activated receptor γ (PPARγ) and insulin-degrading enzyme (IDE) in the hippocampus of rat model of Alzheimer's disease (AD) to determine how ginsenoside Rg1 (Rg1) decreases Aß levels in AD. EXPERIMENTAL APPROACH: Experimental AD was induced in rats by a bilateral injection of 10 µg soluble beta-amyloid peptide 1-42 (Aß(1-42)) into the CA1 region of the hippocampus, and the rats were treated with Rg1 (10 mg·kg(-1), intraperitoneally) for 28 days. The Morris water maze was used to test spatial learning and memory performance. Hematoxylin-eosin staining was performed to analyze the hippocampal histopathological damage. Immunohistochemistry, western blotting, and real-time PCR were used to detect Aß(1-42), PPARγ, and insulin-degrading enzyme (IDE) expression in the hippocampus. KEY RESULTS: Injection of soluble Aß(1-42) into the hippocampus led to significant dysfunction of learning and memory, hippocampal histopathological abnormalities and increased Aß(1-42) levels in the hippocampus. Rg1 treatment significantly improved learning and memory function, attenuated hippocampal histopathological abnormalities, reduced Aß(1-42) levels and increased PPARγ and IDE expression in the hippocampus; these effects of Rg1 could be effectively inhibited by GW9662, a PPARγ antagonist. CONCLUSIONS AND IMPLICATIONS: Given that PPARγ can upregulate IDE expression and IDE can degrade Aß(1-42), these results indicate that Rg1 can increase IDE expression in the hippocampus by upregulating PPARγ, leading to decreased Aß levels, attenuated hippocampal histopathological abnormalities and improved learning and memory in a rat model of AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/toxicity , CA1 Region, Hippocampal/metabolism , Central Nervous System Agents/pharmacology , Gene Expression Regulation/drug effects , Ginsenosides/pharmacology , Insulysin/biosynthesis , Nerve Tissue Proteins/biosynthesis , PPAR gamma/biosynthesis , Peptide Fragments/toxicity , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Anilides/pharmacology , Animals , CA1 Region, Hippocampal/pathology , Learning/drug effects , Male , PPAR gamma/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
8.
CNS Neurol Disord Drug Targets ; 11(8): 1045-51, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23244428

ABSTRACT

We reported previously that geniposide showed neurotrophic and neuroprotective activities with the activation of glucagons-like peptide 1 receptor (GLP-1R) in neurons. The current study was designed to further investigate the protective effect of geniposide on ß-amyloid (Aß)-induced cytotoxicity. Our results showed that pre-incubation with geniposide prevented Aß1₋42-induced cell injury in primary cultured cortical neurons. Geniposide also induced the expression of insulin-degrading enzyme (IDE), a major degrading protease of Aß, in a dose-dependent manner. Moreover, bacitracin, an inhibitor of IDE, and RNAi on Glp-1r gene decreased the neuroprotection of geniposide in Aß1₋42-treated cortical neurons. Our findings indicated that geniposide activating GLP-1R to against Aß-induced neurotoxicity involved in its regulation on the expression of IDE in cortical neurons, which provided an additional mechanistic insight into the role of GLP-1R in neuroprotection.


Subject(s)
Amyloid beta-Peptides/toxicity , Cerebral Cortex/enzymology , Gene Expression Regulation, Enzymologic , Insulysin/biosynthesis , Iridoids/pharmacology , Neurons/enzymology , Peptide Fragments/toxicity , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Dose-Response Relationship, Drug , Female , Male , Neurons/drug effects , Neurons/pathology , Peptide Fragments/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
9.
BMC Med Genet ; 13: 89, 2012 Sep 24.
Article in English | MEDLINE | ID: mdl-22998375

ABSTRACT

BACKGROUND: There is evidence that one of the key type 2 diabetes (T2D) loci identified by GWAS exerts its influence early on in life through its impact on pediatric BMI. This locus on 10q23 harbors three genes, encoding hematopoietically expressed homeobox (HHEX), insulin-degrading enzyme (IDE) and kinesin family member 11 (KIF11), respectively. METHODS: We analyzed the impact of adipogeneis on the mRNA and protein expression levels of these genes in the human adipocyte Simpson-Golabi-Behmel syndrome (SGBS) cell line in order to investigate which could be the culprit gene(s) in this region of linkage disequilibrium. RESULTS: Following activation of differentiation with a PPARγ ligand, we observed ~20% decrease in IDE, ~40% decrease in HHEX and in excess of 80% decrease in KIF11 mRNA levels when comparing the adipocyte and pre-adipocyte states. We also observed decreases in KIF11 and IDE protein levels, but conversely we observed a dramatic increase in HHEX protein levels. Subsequent time course experiments revealed some marked changes in expression as early as three hours after activation of differentiation. CONCLUSION: Our data suggest that the expression of all three genes at this locus are impacted during SGBS adipogenesis and provides insights in to the possible mechanisms of how the genes at this 10q23 locus could influence both adipocyte differentiation and susceptibility to T2D through insulin resistance.


Subject(s)
Body Mass Index , Chromosomes, Human, Pair 10/genetics , Diabetes Mellitus, Type 2/genetics , Gene Expression Profiling , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Adipogenesis/genetics , Arrhythmias, Cardiac/genetics , Cell Line , Chromans/pharmacology , Genetic Diseases, X-Linked/genetics , Genetic Loci , Genetic Predisposition to Disease , Gigantism/genetics , Heart Defects, Congenital/genetics , Homeodomain Proteins/biosynthesis , Humans , Hypoglycemic Agents/pharmacology , Insulin Resistance/genetics , Insulysin/biosynthesis , Intellectual Disability/genetics , Kinesins/biosynthesis , Linkage Disequilibrium/genetics , Rosiglitazone , Thiazolidinediones/pharmacology , Transcription Factors/biosynthesis , Troglitazone
10.
PLoS One ; 7(8): e42035, 2012.
Article in English | MEDLINE | ID: mdl-22870279

ABSTRACT

CpG island promoters often lack canonical core promoter elements such as the TATA box, and have dispersed transcription initiation sites. Despite the prevalence of CpG islands associated with mammalian genes, the mechanism of transcription initiation from CpG island promoters remains to be clarified. Here we investigate the mechanism of transcription initiation of the CpG island-associated gene, insulin-degrading enzyme (IDE). IDE is ubiquitously expressed, and has dispersed transcription initiation sites. The IDE core promoter locates within a 32-bp region, which contains three CGGCG repeats and a nuclear respiratory factor 1 (NRF-1) binding motif. Sequential mutation analysis indicates that the NRF-1 binding motif is critical for IDE transcription initiation. The NRF-1 binding motif is functional, because NRF-1 binds to this motif in vivo and this motif is required for the regulation of IDE promoter activity by NRF-1. Furthermore, the NRF-1 binding site in the IDE promoter is conserved among different species, and dominant negative NRF-1 represses endogenous IDE expression. Finally, TATA-box binding protein (TBP) is not associated with the IDE promoter, and inactivation of TBP does not abolish IDE transcription, suggesting that TBP is not essential for IDE transcription initiation. Our studies indicate that NRF-1 mediates IDE transcription initiation in a TBP-independent manner, and provide insights into the potential mechanism of transcription initiation for other CpG island-associated genes.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Insulysin/biosynthesis , Nuclear Respiratory Factor 1/metabolism , TATA Box/physiology , Transcription Initiation, Genetic/physiology , Animals , CpG Islands/physiology , HeLa Cells , Humans , Insulysin/genetics , Mice , NIH 3T3 Cells , Nuclear Respiratory Factor 1/genetics , Response Elements/physiology
11.
Hum Mol Genet ; 21(13): 2973-90, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22492996

ABSTRACT

The purpose of this study was to investigate the protective effects of the mitochondria-targeted antioxidant catalase (MCAT) and lifespan extension in mice that express amyloid beta (Aß). Using immunoblotting and immunostaining analyses, we measured the production of full-length amyloid precursor protein (APP), soluble APPα, C-terminal fragments CTF99 and CTF83, monomeric and oligomeric Aß, Aß deposits and beta site amyloid precursor protein cleaving enzyme 1 (BACE1), in different stages of disease progression in MCAT/AßPP and AßPP mice. Using quantitative reverse transcriptase polymerase chain reaction and immunostaining analyses, we studied the expression of catalase, BACE1, the Alzheimer's disease (AD) markers, synaptophysin, APP, neprilysin, insulin-degrading enzyme and transthyretin in MCAT, AßPP, MCAT/AßPP and wild-type (WT) mice. Using the high pressure liquid chromatography analysis of 8-hydroxy-2-deoxyguanosine, we measured oxidative DNA damage in the cerebral cortical tissues from MCAT, AßPP, MCAT/AßPP and WT mice. We found that the AßPP transgenic mice that carried the human MCAT gene lived 5 months longer than did the AßPP mice. We also found that the overexpression of MCAT in the brain sections from the MCAT/AßPP transgenic mice significantly correlated with a reduction in the levels of full-length APP, CTF99, BACE1, Aß levels (40 and 42), Aß deposits and oxidative DNA damage relative to the brain sections from the AßPP mice. Interestingly, we found significantly increased levels of soluble APPα and CTF83 in the MCAT/AßPP mice, relative to the AßPP mice. These data provide direct evidence that oxidative stress plays a primary role in AD etiopathology and that in MCAT mice express Aß, MCAT prevents abnormal APP processing, reduces Aß levels and enhances Aß-degrading enzymes in mice at different ages, corresponding to different stages of disease progression. These findings indicate that mitochondria-targeted molecules may be an effective therapeutic approach to treat patients with AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/biosynthesis , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/biosynthesis , Catalase/metabolism , Mitochondria/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/biosynthesis , Animals , Brain/pathology , Catalase/genetics , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , DNA Damage/genetics , Disease Models, Animal , Female , Insulysin/biosynthesis , Insulysin/metabolism , Male , Mice , Mice, Transgenic , Neprilysin/biosynthesis , Neuroprotective Agents/metabolism , Oxidative Stress , Prealbumin/biosynthesis , RNA, Messenger/biosynthesis , Random Allocation , Synaptophysin/biosynthesis
12.
Diabetologia ; 55(5): 1495-504, 2012 May.
Article in English | MEDLINE | ID: mdl-22349107

ABSTRACT

AIMS/HYPOTHESIS: Ciliary neurotrophic factor (CNTF) improves metabolic variables of obese animals with characteristics of type 2 diabetes, mainly by reducing insulin resistance. We evaluated whether CNTF was able to improve other metabolic variables in mouse models of type 2 diabetes, such as beta cell mass and insulin clearance, and whether CNTF has any effect on non-obese mice with characteristics of type 2 diabetes. METHODS: Neonatal mice were treated with 0.1 mg/kg CNTF or citrate buffer via intraperitoneal injections, before injection of 250 mg/kg alloxan. HEPG2 cells were cultured for 3 days in the presence of citrate buffer, 1 nmol/l CNTF or 50 mmol/l alloxan or a combination of CNTF and alloxan. Twenty-one days after treatment, we determined body weight, epididymal fat weight, blood glucose, plasma insulin, NEFA, glucose tolerance, insulin resistance, insulin clearance and beta cell mass. Finally, we assessed insulin receptor and protein kinase B phosphorylation in peripheral organs, as well as insulin-degrading enzyme (IDE) protein production and alternative splicing in the liver and HEPG2 cells. RESULTS: CNTF improved insulin sensitivity and beta cell mass, while reducing glucose-stimulated insulin secretion and insulin clearance in Swiss mice, improving glucose handling in a non-obese type 2 diabetes model. This effect was associated with lower IDE production and activity in liver cells. All these effects were observed even at 21 days after CNTF treatment. CONCLUSIONS/INTERPRETATION: CNTF protection against type 2 diabetes is partially independent of the anti-obesity actions of CNTF, requiring a reduction in insulin clearance and increased beta cell mass, besides increased insulin sensitivity. Furthermore, knowledge of the long-term effects of CNTF expands its pharmacological relevance.


Subject(s)
Ciliary Neurotrophic Factor/pharmacology , Diabetes Mellitus, Experimental/prevention & control , Diabetes Mellitus, Type 2/prevention & control , Insulin-Secreting Cells/drug effects , Insulin/metabolism , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Diabetes Mellitus, Type 2/metabolism , Epididymis/drug effects , Fatty Acids, Nonesterified/blood , Hep G2 Cells , Humans , Insulin/blood , Insulin Resistance , Insulin Secretion , Insulysin/biosynthesis , Male , Mice , Proto-Oncogene Proteins c-akt/analysis , Receptor, Insulin/analysis
13.
J Neurosci ; 30(35): 11848-57, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20810904

ABSTRACT

Locus ceruleus (LC) is the main subcortical site of norepinephrine synthesis. In Alzheimer's disease (AD) patients and rodent models, degeneration of LC neurons and reduced levels of norepinephrine in LC projection areas are significantly correlated with the increase in amyloid plaques, neurofibrillary tangles, and severity of dementia. Activated microglia play a pivotal role in the progression of AD by either clearing amyloid beta peptide (Abeta) deposits through uptake of Abeta or releasing cytotoxic substances and proinflammatory cytokines. Here, we investigated the effect of norepinephrine on Abeta uptake and clearance by murine microglia and explored the underlying mechanisms. We found that murine microglia cell line N9 and primary microglia expressed beta(2) adrenergic receptor (AR) but not beta(1) and beta(3)AR. Norepinephrine and isoproterenol upregulated the expression of Abeta receptor mFPR2, a mouse homolog of human formyl peptide receptor FPR2, through activation of beta(2)AR in microglia. Norepinephrine also induced mFPR2 expression in mouse brain. Activation of beta(2)AR in microglia promoted Abeta(42) uptake through upregulation of mFPR2 and enhanced spontaneous cell migration but had no effect on cell migration in response to mFPR2 agonists. Furthermore, activation of beta(2)AR on microglia induced the expression of insulin-degrading enzyme and increased the degradation of Abeta(42). Mechanistic studies showed that isoproterenol induced mFPR2 expression through ERK1/2-NF-kappaB and p38-NF-kappaB signaling pathways. These findings suggest that noradrenergic innervation from LC is needed to maintain adequate Abeta uptake and clearance by microglia, and norepinephrine is a link between neuron and microglia to orchestrate the host response to Abeta in AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Insulysin/biosynthesis , Microglia/metabolism , Norepinephrine/pharmacology , Peptide Fragments/metabolism , Receptors, Formyl Peptide/biosynthesis , Up-Regulation/physiology , Adrenergic beta-Agonists/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Animals, Newborn , Cell Line , Cells, Cultured , Endocytosis/drug effects , Endocytosis/physiology , Humans , Insulysin/physiology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Norepinephrine/physiology , Peptide Fragments/antagonists & inhibitors , Receptors, Formyl Peptide/physiology , Up-Regulation/drug effects
14.
J Antibiot (Tokyo) ; 63(9): 549-52, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20606694

ABSTRACT

Two peptides, tumescenamides A and B, were isolated from the fermentation broth of a marine bacterium, Streptomyces tumescens YM23-260. The structure of tumescenamide A was determined to be a cyclic depsipeptide consisting of α-amino-2-butenoic acid, tyrosine, valine, leucine and threonine, substituted with a 2,4-dimethylheptanoyl residue at the α-NH(2) position. Tumescenamide B possesses a 2,4,6-trimethylnonanoyl residue in place of the 2,4-dimethylheptanoyl substituent in tumescenamide A. Tumescenamide A induced reporter gene expression under the control of the insulin-degrading enzyme promoter.


Subject(s)
Depsipeptides/chemistry , Depsipeptides/isolation & purification , Streptomyces/metabolism , Amino Acids/analysis , Depsipeptides/metabolism , Depsipeptides/pharmacology , Genes, Reporter , Insulysin/biosynthesis , Insulysin/genetics , Luciferases/biosynthesis , Luciferases/genetics , Magnetic Resonance Spectroscopy , Molecular Structure , Promoter Regions, Genetic , Transcriptional Activation
15.
J Neurochem ; 115(1): 47-57, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20663017

ABSTRACT

The brain steady state level of ß-amyloid (Aß) is determined by the balance between its production and removal, the latter through egress across blood and CSF barriers as well as Aß degradation. The major Aß-degrading enzymes are neprilysin (NEP), insulin-degrading enzyme (IDE), and endothelin-converting enzyme (ECE-1). Although evidence suggests that NEP is down-regulated in Alzheimer's disease (AD), the role of IDE and ECE in the Aß accumulation in aging and dementia remains less certain. In this study, we examined mRNA and protein expression, as well as biological activity of NEP, IDE, and ECE-1 in human frontal cortex by real-time RT-PCR for mRNA, immunoblotting for protein, and highly sensitive and specific fluorescence assays for activity. The relationships between Aß-degrading enzymes and pathologic measures and clinical features were also assessed. The results showed that NEP mRNA, protein level, and activity were decreased in AD compared with normal controls with no cognitive impairment (NCI). In contrast, IDE activity was unchanged, but there was higher expression of IDE mRNA, indicating a possible compensatory reaction because of deficits in activity. ECE-1 expression in AD brain showed no significant difference compared with age-matched controls. Correlation analyses suggested that NEP expression was correlated with Aß accumulation and clinical diagnosis, being lower in AD than in no cognitive impairment. In contrast, neither IDE nor ECE-1 correlated with Aß or clinical diagnosis. These findings provide additional support for NEP as the major protease involved in Aß degradation and suggest its possible therapeutic targeting in AD.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Aspartic Acid Endopeptidases/biosynthesis , Aspartic Acid Endopeptidases/genetics , Brain Chemistry/genetics , Insulysin/biosynthesis , Insulysin/genetics , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/genetics , Neprilysin/biosynthesis , Neprilysin/genetics , Aged , Aged, 80 and over , Alzheimer Disease/psychology , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/genetics , Blotting, Western , Brain/enzymology , Cognition Disorders/enzymology , Cognition Disorders/genetics , DNA Primers , Electrophoresis, Polyacrylamide Gel , Endothelin-Converting Enzymes , Female , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/physiology , Humans , Indicators and Reagents , Male , Prospective Studies , RNA/biosynthesis , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Synaptophysin/biosynthesis , Synaptophysin/genetics , Tissue Banks
16.
Pharmazie ; 65(3): 231-2, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20383947

ABSTRACT

Previous results show that treatment with saturated fatty acids, such as palmitic acid (PA), induces the pathology of Alzheimer's disease (AD), while treatment with polyunsaturated fatty acids, such as docosahexaenoic acid (DHA), protects against AD pathology. However, the pharmacological mechanism underlying these opposite effects of fatty acids on AD is not well understood. Here, we show that PA treatment significantly reduced the expression of insulin-degrading enzyme (IDE), an important protease responsible for the degradation of amyloid-beta (A beta) in neural cells, while incubation with DHA up-regulated IDE levels in primary hippocampal neurons. Moreover, pre-incubation with PA attenuated the DHA-induced IDE expression. Taken together, these results suggest the opposite effects of saturated fatty acids and polyunsaturated fatty acids on the expression of IDE, indicating a novel mechanism underlying the pharmacological function of fatty acids in AD intervention.


Subject(s)
Docosahexaenoic Acids/pharmacology , Enzyme Inhibitors/pharmacology , Insulin/metabolism , Insulysin/biosynthesis , Neurons/drug effects , Neurons/enzymology , Palmitic Acid/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Animals , Blotting, Western , Cells, Cultured , Gene Expression Regulation, Enzymologic/drug effects , Insulysin/genetics , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
17.
Stress ; 13(2): 123-31, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19929311

ABSTRACT

We investigated whether long-term administration of exogenous corticosterone (CST) or vehicle as daily treatment induces changes in rat behavior and in gene expression of the rat brain insulin signaling pathway and the formation of tau protein. Two groups of male adult rats received daily subcutaneous injections of 26.8 mg/kg CST (CST stress group) or vehicle-sesame oil (injection stress group) for 60 days while the third group was taken as untreated controls (n = 8 each). Body weight and plasma CST were measured and psychometric investigations were conducted using a rat holeboard test system before and after the treatment. Gene expression analyzes were performed by RT-PCR in cerebral cortical tissue for insulin genes 1 and 2, insulin receptor (IR), insulin degrading enzyme (IDE), and tau protein. Daily injections of CST for 60 days induced a significant, 2-fold increase in rat plasma CST concentrations in comparison to untreated controls. Significantly reduced behavioral abilities in CST-treated rats were associated with reduced gene expression of insulin 1 ( - 20%), IDE ( - 23%), and IR ( - 26%), indicating an insulin-resistant brain state, followed by increased tau protein (+28%) gene expression. In summary, chronic CST administration affects gene expression in the brain IR signaling cascade and increases tau gene expression, which is associated with reductions in cognition capacity in rats.


Subject(s)
Cerebral Cortex/drug effects , Corticosterone/pharmacology , Insulin Resistance/physiology , Animals , Behavior, Animal/drug effects , Cognition Disorders/chemically induced , Corticosterone/blood , Gene Expression/drug effects , Insulin/biosynthesis , Insulysin/biosynthesis , Male , Memory/drug effects , Rats , Rats, Wistar , Receptor, Insulin/biosynthesis , Stress, Psychological/physiopathology , Stress, Psychological/psychology , tau Proteins/biosynthesis
18.
PLoS One ; 4(10): e7643, 2009 Oct 28.
Article in English | MEDLINE | ID: mdl-19862325

ABSTRACT

BACKGROUND: Recent advance in genetic studies added the confirmed susceptible loci for type 2 diabetes to eighteen. In this study, we attempt to analyze the independent and joint effect of variants from these loci on type 2 diabetes and clinical phenotypes related to glucose metabolism. METHODS/PRINCIPAL FINDINGS: Twenty-one single nucleotide polymorphisms (SNPs) from fourteen loci were successfully genotyped in 1,849 subjects with type 2 diabetes and 1,785 subjects with normal glucose regulation. We analyzed the allele and genotype distribution between the cases and controls of these SNPs as well as the joint effects of the susceptible loci on type 2 diabetes risk. The associations between SNPs and type 2 diabetes were examined by logistic regression. The associations between SNPs and quantitative traits were examined by linear regression. The discriminative accuracy of the prediction models was assessed by area under the receiver operating characteristic curves. We confirmed the effects of SNPs from PPARG, KCNJ11, CDKAL1, CDKN2A-CDKN2B, IDE-KIF11-HHEX, IGF2BP2 and SLC30A8 on risk for type 2 diabetes, with odds ratios ranging from 1.114 to 1.406 (P value range from 0.0335 to 1.37E-12). But no significant association was detected between SNPs from WFS1, FTO, JAZF1, TSPAN8-LGR5, THADA, ADAMTS9, NOTCH2-ADAM30 and type 2 diabetes. Analyses on the quantitative traits in the control subjects showed that THADA SNP rs7578597 was association with 2-h insulin during oral glucose tolerance tests (P = 0.0005, empirical P = 0.0090). The joint effect analysis of SNPs from eleven loci showed the individual carrying more risk alleles had a significantly higher risk for type 2 diabetes. And the type 2 diabetes patients with more risk allele tended to have earlier diagnostic ages (P = 0.0006). CONCLUSIONS/SIGNIFICANCE: The current study confirmed the association between PPARG, KCNJ11, CDKAL1, CDKN2A-CDKN2B, IDE-KIF11-HHEX, IGF2BP2 and SLC30A8 and type 2 diabetes. These type 2 diabetes risk loci contributed to the disease additively.


Subject(s)
Cation Transport Proteins/biosynthesis , Cyclin-Dependent Kinase 5/biosynthesis , Cyclin-Dependent Kinase Inhibitor p15/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Diabetes Mellitus, Type 2/ethnology , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Homeodomain Proteins/biosynthesis , Insulysin/biosynthesis , Kinesins/biosynthesis , PPAR gamma/biosynthesis , Potassium Channels, Inwardly Rectifying/biosynthesis , RNA-Binding Proteins/biosynthesis , Transcription Factors/biosynthesis , Aged , China , Female , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Zinc Transporter 8 , tRNA Methyltransferases
19.
Age (Dordr) ; 31(4): 277-84, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19468866

ABSTRACT

The purpose of this study was to look for alterations in ß-amyloid peptide (Aß) metabolism-related molecules in predementia, the early stage of Alzheimer's disease (AD). AlCl3 (Al) and d-galactose (D-gal) were used to induce the mouse model for predementia and AD. Protein expression of ß-amyloid (Aß), ß-secretase (BACE1), neprilysin (NEP), insulin degrading enzyme (IDE) and receptor for advanced glycation end products (RAGE) in the brain was measured. The results indicated that Al + D-gal induced an AD-like behavioral deficit at 90 days. The period from 45 to 75 days showed no significant behavioral deficit, and we tentatively define this as predementia in this model. A significant increase in BACE1 and decreasing NEP characterized days 45­90 in the cortex and hippocampus. However, high Aß occurred at day 60. IDE increased from day 60 to day 75. There was no change in RAGE. The results suggest that the observed changes in BACE1, NEP and Aß in predementia might relate to a different stage of the AD-like pathology, which may be developed into useful biomarkers for the diagnosis of very early AD.


Subject(s)
Aluminum Compounds/pharmacology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Cerebral Cortex/metabolism , Chlorides/pharmacology , Galactose/pharmacology , Hippocampus/metabolism , Aluminum Chloride , Alzheimer Disease/diagnosis , Alzheimer Disease/prevention & control , Amyloid Precursor Protein Secretases/biosynthesis , Animals , Biomarkers/metabolism , Blotting, Western , Disease Models, Animal , Disease Progression , Follow-Up Studies , Glycation End Products, Advanced/metabolism , Hippocampus/drug effects , Insulysin/biosynthesis , Mice , Mice, Transgenic , Neprilysin/biosynthesis
20.
J Immunol ; 181(9): 6503-13, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941241

ABSTRACT

A hallmark of immunopathology associated with Alzheimer's disease is the presence of activated microglia (MG) surrounding senile plaque deposition of beta-amyloid (Abeta) peptides. Abeta peptides are believed to be potent activators of MG, which leads to Alzheimer's disease pathology, but the role of MG subtypes in Abeta clearance still remains unclear. In this study, we found that IL-4 treatment of rat primary-type 2 MG enhanced uptake and degradation of oligomeric Abeta(1-42) (o-Abeta(1-42)). IL-4 treatment induced significant expression of the scavenger receptor CD36 and the Abeta-degrading enzymes neprilysin (NEP) and insulin-degrading enzyme (IDE) but reduced expression of certain other scavenger receptors. Of cytokines and stimulants tested, the anti-inflammatory cytokines IL-4 and IL-13 effectively enhanced CD36, NEP, and IDE. We demonstrated the CD36 contribution to IL-4-induced Abeta clearance: Chinese hamster ovary cells overexpressing CD36 exhibited marked, dose-dependent degradation of (125)I-labeled o-Abeta(1-42) compared with controls, the degradation being blocked by anti-CD36 Ab. Also, we found IL-4-induced clearance of o-Abeta(1-42) in type 2 MG from CD36-expressing WKY/NCrj rats but not in cells from SHR/NCrj rats with dysfunctional CD36 expression. NEP and IDE also contributed to IL-4-induced degradation of Abeta(1-42), because their inhibitors, thiorphan and insulin, respectively, significantly suppressed this activity. IL-4-stimulated uptake and degradation of o-Abeta(1-42) were selectively enhanced in type 2, but not type 1 MG that express CD40, which suggests that the two MG types may play different neuroimmunomodulating roles in the Abeta-overproducing brain. Thus, selective o-Abeta(1-42) clearance, which is induced by IL-4, may provide an additional focus for developing strategies to prevent and treat Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Interleukin-4/physiology , Microglia/immunology , Microglia/metabolism , Peptide Fragments/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Amyloid beta-Peptides/antagonists & inhibitors , Animals , CD36 Antigens/biosynthesis , CD36 Antigens/genetics , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Dimerization , Humans , Insulysin/biosynthesis , Insulysin/genetics , Lysosomal Membrane Proteins/biosynthesis , Lysosomal Membrane Proteins/genetics , Mice , Microglia/classification , Microglia/pathology , Neprilysin/biosynthesis , Neprilysin/genetics , Peptide Fragments/antagonists & inhibitors , Rats , Rats, Wistar , Receptors, Scavenger/biosynthesis , Receptors, Scavenger/genetics , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...