Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
1.
Proc Natl Acad Sci U S A ; 117(33): 20149-20158, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32747560

ABSTRACT

The C2 domain containing protein extended synaptotagmin (E-Syt) plays important roles in both lipid homeostasis and the intracellular signaling; however, its role in physiology remains largely unknown. Here, we show that hypothalamic E-Syt3 plays a critical role in diet-induced obesity (DIO). E-Syt3 is characteristically expressed in the hypothalamic nuclei. Whole-body or proopiomelanocortin (POMC) neuron-specific ablation of E-Syt3 ameliorated DIO and related comorbidities, including glucose intolerance and dyslipidemia. Conversely, overexpression of E-Syt3 in the arcuate nucleus moderately promoted food intake and impaired energy expenditure, leading to increased weight gain. Mechanistically, E-Syt3 ablation led to increased processing of POMC to α-melanocyte-stimulating hormone (α-MSH), increased activities of protein kinase C and activator protein-1, and enhanced expression of prohormone convertases. These findings reveal a previously unappreciated role for hypothalamic E-Syt3 in DIO and related metabolic disorders.


Subject(s)
Gene Expression Regulation/physiology , Obesity/chemically induced , Obesity/genetics , Synaptotagmins/metabolism , Animals , Diet, High-Fat/adverse effects , Genetic Predisposition to Disease , Hypothalamus/metabolism , Male , Mice , Mice, Knockout , Neurons/metabolism , Proprotein Convertase 1/genetics , Proprotein Convertase 1/metabolism , Proprotein Convertase 2/genetics , Proprotein Convertase 2/metabolism , Synaptotagmins/genetics
2.
J Biol Chem ; 297(1): 100839, 2021 07.
Article in English | MEDLINE | ID: mdl-34051232

ABSTRACT

Glucose-mediated signaling regulates the expression of a limited number of genes in human pancreatic ß-cells at the transcriptional level. However, it is unclear whether glucose plays a role in posttranscriptional RNA processing or translational control of gene expression. Here, we asked whether glucose affects posttranscriptional steps and regulates protein synthesis in human ß-cell lines. We first showed the involvement of the mTOR pathway in glucose-related signaling. We also used the surface sensing of translation technique, based on puromycin incorporation into newly translated proteins, to demonstrate that glucose treatment increased protein translation. Among the list of glucose-induced proteins, we identified the proconvertase PCSK1, an enzyme involved in the proteolytic conversion of proinsulin to insulin, whose translation was induced within minutes following glucose treatment. We finally performed global proteomic analysis by mass spectrometry to characterize newly translated proteins upon glucose treatment. We found enrichment in proteins involved in translation, glycolysis, TCA metabolism, and insulin secretion. Taken together, our study demonstrates that, although glucose minorly affects gene transcription in human ß-cells, it plays a major role at the translational level.


Subject(s)
Energy Metabolism/genetics , Glucose/pharmacology , Insulin Secretion/genetics , Insulin-Secreting Cells/metabolism , Protein Biosynthesis/genetics , Cell Line , Cyclic AMP-Dependent Protein Kinase RIIalpha Subunit/metabolism , Energy Metabolism/drug effects , Humans , Insulin Secretion/drug effects , Insulin-Secreting Cells/drug effects , Mitogen-Activated Protein Kinases/metabolism , Proprotein Convertase 1/metabolism , Protein Biosynthesis/drug effects , Puromycin/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
3.
Mol Cell ; 56(3): 425-435, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25458843

ABSTRACT

Pyruvate lies at a central biochemical node connecting carbohydrate, amino acid, and fatty acid metabolism, and the regulation of pyruvate flux into mitochondria represents a critical step in intermediary metabolism impacting numerous diseases. To characterize changes in mitochondrial substrate utilization in the context of compromised mitochondrial pyruvate transport, we applied (13)C metabolic flux analysis (MFA) to cells after transcriptional or pharmacological inhibition of the mitochondrial pyruvate carrier (MPC). Despite profound suppression of both glucose and pyruvate oxidation, cell growth, oxygen consumption, and tricarboxylic acid (TCA) metabolism were surprisingly maintained. Oxidative TCA flux was achieved through enhanced reliance on glutaminolysis through malic enzyme and pyruvate dehydrogenase (PDH) as well as fatty acid and branched-chain amino acid oxidation. Thus, in contrast to inhibition of complex I or PDH, suppression of pyruvate transport induces a form of metabolic flexibility associated with the use of lipids and amino acids as catabolic and anabolic fuels.


Subject(s)
Proprotein Convertase 1/metabolism , Proprotein Convertase 2/metabolism , Pyruvic Acid/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Citric Acid Cycle , Fatty Acids/metabolism , Glutamine/metabolism , Humans , Lipogenesis , Metabolic Flux Analysis , Mice , Muscle Fibers, Skeletal/metabolism , Oxidation-Reduction
4.
Int J Mol Sci ; 23(15)2022 Aug 05.
Article in English | MEDLINE | ID: mdl-35955861

ABSTRACT

Prader−Willi syndrome (PWS) is a complex neurodevelopmental disorder caused by the deletion or inactivation of paternally expressed imprinted genes at the chromosomal region 15q11−q13. The PWS-critical region (PWScr) harbors tandemly repeated non-protein coding IPW-A exons hosting the intronic SNORD116 snoRNA gene array that is predominantly expressed in brain. Paternal deletion of PWScr is associated with key PWS symptoms in humans and growth retardation in mice (PWScr model). Dysregulation of the hypothalamic−pituitary axis (HPA) is thought to be causally involved in the PWS phenotype. Here we performed a comprehensive reverse transcription quantitative PCR (RT-qPCR) analysis across nine different brain regions of wild-type (WT) and PWScr mice to identify stably expressed reference genes. Four methods (Delta Ct, BestKeeper, Normfinder and Genorm) were applied to rank 11 selected reference gene candidates according to their expression stability. The resulting panel consists of the top three most stably expressed genes suitable for gene-expression profiling and comparative transcriptome analysis of WT and/or PWScr mouse brain regions. Using these reference genes, we revealed significant differences in the expression patterns of Igfbp7, Nlgn3 and three HPA associated genes: Pcsk1, Pcsk2 and Nhlh2 across investigated brain regions of wild-type and PWScr mice. Our results raise a reasonable doubt on the involvement of the Snord116 in posttranscriptional regulation of Nlgn3 and Nhlh2 genes. We provide a valuable tool for expression analysis of specific genes across different areas of the mouse brain and for comparative investigation of PWScr mouse models to discover and verify different regulatory pathways affecting this complex disorder.


Subject(s)
Prader-Willi Syndrome , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/metabolism , Disease Models, Animal , Exons , Genomic Imprinting , Humans , Mice , Prader-Willi Syndrome/genetics , Prader-Willi Syndrome/metabolism , Proprotein Convertase 1/genetics , Proprotein Convertase 1/metabolism , RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism
5.
Biochem Biophys Res Commun ; 526(3): 618-625, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32248978

ABSTRACT

Glucose mediated insulin biosynthesis is tightly regulated and shared between insulin granule proteins such as its processing enzymes, prohormone convertases, PC1/3 and PC2. However, the molecular players involved in the co-ordinated translation remain elusive. The trans-acting factors like PABP (Poly A Binding Protein) and PDI (Protein Disulphide Isomerize) binds to a conserved sequence in the 5'UTR of insulin mRNA and regulates its translation. Here, we demonstrate that 5'UTR of PC1/3 and PC2 also associate with PDI and PABP. We show that a' and RRM 3-4 domains of PDI and PABP respectively, are necessary for RNA binding activity to the 5'UTRs of insulin and its processing enzymes.


Subject(s)
Insulin/metabolism , Poly(A)-Binding Proteins/metabolism , Proprotein Convertase 1/metabolism , Proprotein Convertase 2/metabolism , Protein Biosynthesis , Protein Disulfide-Isomerases/metabolism , 5' Untranslated Regions , Animals , Cell Line , Cytoplasmic Granules/genetics , Cytoplasmic Granules/metabolism , Insulin/genetics , Mice , Poly(A)-Binding Proteins/genetics , Proprotein Convertase 1/genetics , Proprotein Convertase 2/genetics , Protein Disulfide-Isomerases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Mamm Genome ; 31(1-2): 17-29, 2020 02.
Article in English | MEDLINE | ID: mdl-31974728

ABSTRACT

The proprotein convertase subtilisin/Kexin type 1 (PCSK1/PC1) protein processes inactive pro-hormone precursors into biologically active hormones in a number of neuroendocrine and endocrine cell types. Patients with recessive mutations in PCSK1 exhibit a complex spectrum of traits including obesity, diarrhoea and endocrine disorders. We describe here a new mouse model with a point mutation in the Pcsk1 gene that exhibits obesity, hyperphagia, transient diarrhoea and hyperproinsulinaemia, phenotypes consistent with human patient traits. The mutation results in a pV96L amino acid substitution and changes the first nucleotide of mouse exon 3 leading to skipping of that exon and in homozygotes very little full-length transcript. Overexpression of the exon 3 deleted protein or the 96L protein results in ER retention in Neuro2a cells. This is the second Pcsk1 mouse model to display obesity phenotypes, contrasting knockout mouse alleles. This model will be useful in investigating the basis of endocrine disease resulting from prohormone processing defects.


Subject(s)
Diabetes Mellitus/genetics , Obesity/genetics , Proprotein Convertase 1/genetics , Alleles , Animals , Cell Line , Diarrhea/genetics , Endoplasmic Reticulum/metabolism , Exons , Female , Glucose Intolerance/genetics , Homozygote , Hyperphagia/genetics , Male , Mice , Mutation , Proprotein Convertase 1/metabolism , RNA Splicing
7.
Diabetologia ; 61(2): 284-294, 2018 02.
Article in English | MEDLINE | ID: mdl-28956082

ABSTRACT

AIMS/HYPOTHESIS: Enteroendocrine K and L cells are pivotal in regulating appetite and glucose homeostasis. Knowledge of their distribution in humans is sparse and it is unknown whether alterations occur in type 2 diabetes. We aimed to evaluate the distribution of enteroendocrine K and L cells and relevant prohormone-processing enzymes (using immunohistochemical staining), and to evaluate the mRNA expression of the corresponding genes along the entire intestinal tract in individuals with type 2 diabetes and healthy participants. METHODS: In this cross-sectional study, 12 individuals with type 2 diabetes and 12 age- and BMI-matched healthy individuals underwent upper and lower double-balloon enteroscopy with mucosal biopsy retrieval from approximately every 30 cm of the small intestine and from seven specific anatomical locations in the large intestine. RESULTS: Significantly different densities for cells positive for chromogranin A (CgA), glucagon-like peptide-1, glucose-dependent insulinotropic polypeptide, peptide YY, prohormone convertase (PC) 1/3 and PC2 were observed along the intestinal tract. The expression of CHGA did not vary along the intestinal tract, but the mRNA expression of GCG, GIP, PYY, PCSK1 and PCSK2 differed along the intestinal tract. Lower counts of CgA-positive and PC1/3-positive cells, respectively, were observed in the small intestine of individuals with type 2 diabetes compared with healthy participants. In individuals with type 2 diabetes compared with healthy participants, the expression of GCG and PYY was greater in the colon, while the expression of GIP and PCSK1 was greater in the small intestine and colon, and the expression of PCSK2 was greater in the small intestine. CONCLUSIONS/INTERPRETATION: Our findings provide a detailed description of the distribution of enteroendocrine K and L cells and the expression of their products in the human intestinal tract and demonstrate significant differences between individuals with type 2 diabetes and healthy participants. TRIAL REGISTRATION: NCT03044860.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Enteroendocrine Cells/metabolism , Adult , Aged , Chromogranin A/metabolism , Cross-Sectional Studies , Female , Gastric Inhibitory Polypeptide/metabolism , Gastrointestinal Tract/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Peptide YY/metabolism , Proprotein Convertase 1/metabolism , Proprotein Convertase 2/metabolism , Proprotein Convertases/metabolism
8.
J Cell Physiol ; 233(9): 6952-6964, 2018 09.
Article in English | MEDLINE | ID: mdl-29336483

ABSTRACT

In previous studies on glucose metabolism during in vitro maturation, intact cumulus-oocyte complexes (COCs) were treated with enzyme inhibitors/activators. Because inhibitors/activators may have non-specificity and/or toxicity, and culture of COCs cannot differentiate whether glucose metabolism of cumulus cells (CCs) or that of the oocyte supports oocyte maturation, results from the previous studies must be verified by silencing genes in either CCs or cumulus-denuded oocytes (DOs). In this study, RNAi was adopted to specify the effects of glucose metabolism in CCs or DOs on oocyte maturation. Although silencing either glyceraldehyde 3-phosphate dehydrogenase (GAPDH) or glucose-6-phosphate dehydrogenase (G6PD) genes in CCs significantly decreased competence of the cocultured DOs, silencing G6PD impaired competence to a greater extent. While silencing G6PD or GAPDH of CCs decreased glutathione and ATP contents of cocultured DOs to similar extents, silencing G6PD increased oxidative stress as well. Analysis on metabolite contents and oxidative stress index and culture of DOs in medium conditioned with gene-silenced CCs indicated that CCs supported oocyte maturation by releasing glucose metabolites. Silencing mitochondrial pyruvate carrier 1 or NADH dehydrogenase (ubiquintone) flavoprotein 1 of DOs significantly impaired their maturation. The results have unequivocally confirmed that CCs promote oocyte maturation by releasing glucose metabolites from both pentose phosphate pathway (PPP) and glycolysis. Pyruvate is transferred into DOs by mitochondrial pyruvate carrier (MPC) and utilized through mitochondrial electron transport to support maturation.


Subject(s)
Glucose/metabolism , In Vitro Oocyte Maturation Techniques , Oocytes/cytology , Oocytes/metabolism , RNA Interference , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cumulus Cells/cytology , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Down-Regulation/drug effects , Electron Transport Complex I/metabolism , Energy Metabolism/drug effects , Glucosephosphate Dehydrogenase/metabolism , Glutathione/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Glycolysis/drug effects , Mice , NADP/metabolism , Oocytes/drug effects , Oxidation-Reduction , Pentose Phosphate Pathway/drug effects , Proprotein Convertase 1/metabolism , Pyruvic Acid/metabolism , Reactive Oxygen Species/metabolism
9.
Biochem Biophys Res Commun ; 495(1): 1008-1013, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29175325

ABSTRACT

Pyruvate is a central substrate in energy metabolism, paramount to carbohydrate, fat, and amino acid catabolic and anabolic pathways. Mitochondrial pyruvate carrier 1(MPC1) is one important component of the complex that facilitates mitochondrial pyruvate import. Complete MPC1 deficiency is a serious concern, and has been shown to result in embryonic lethality in mice. The study outlined in this paper generated one mouse line with the MPC1 protein part deficiency by using the CRISPR/Cas9 system. Clinical observations, body weight and organ/tissue weight, gas exchange, cold-stimulation, blood parameters, as well as histopathology analysis were analyzed to evaluate potential physiological abnormalities caused by MPC1 deficiency. Results indicate that MPC1+/- mice experienced a change in important clinical criteria such as low body weight, decreased movement, and low body shell temperature, few adipose accumulate. The mice show significant difference in some blood parameters including apo-B100, apo-A1, HDL, glucagon, insulin. However these changes alleviated while being fed with the HFD, which provided metabolites to sustain the TCA cycle and body development. The MPC1+/- mice may employ fatty acid oxidation to meet their bioenergetic demands. This study suggests that inhibition of MPC1 activity can boost fatty acid oxidation to provide sufficient energy to the body. This work promotes further studies regarding the interplay between carbohydrate and fat metabolism.


Subject(s)
Body Weight/physiology , Energy Metabolism/physiology , Fatty Acids/metabolism , Oxygen Consumption/physiology , Proprotein Convertase 1/metabolism , Pyruvic Acid/metabolism , Animals , Cold-Shock Response/physiology , Enzyme Activation , Male , Mice , Mice, Knockout , Oxidation-Reduction
10.
Med Sci Monit ; 24: 7614-7624, 2018 Oct 25.
Article in English | MEDLINE | ID: mdl-30356033

ABSTRACT

BACKGROUND Recent studies have illustrated that the transcription co-repressor, C-terminal binding protein 1 (CtBP1), links the metabolic alterations to transcription controls in proliferation, EMT, genome stability, metabolism, and lifespan, but whether CtBP1 affects the cellular redox homeostasis is unexplored. This study was designed to investigate the mechanism of CtBP1-mediated transcription repression that contributes to the metabolic reprogramming. MATERIAL AND METHODS Knockdown of CtBP1 in both mouse MEF cells and human melanoma cells changed cell redox homeostasis. Further, chromatin immunoprecipitation (ChIP) and luciferase reporter assay were performed for identification of CtBP1 downstream targets, pyruvate carrier 1 and 2 genes (MPC1 and MPC2), which contribute to redox homeostasis and are transcriptionally regulated by CtBP1. Moreover, blockage of the cellular NADH level with the glycolysis inhibitor 2-Deoxy-D-Glucose (2-DG) rescued MPC1 and MPC2 expression. MTT assay and scratch assay were performed to investigate the effect of MPC1 and MPC2 expression on malignant properties of melanoma cells. RESULTS The data demonstrated that CtBP1 directly bound to the promoters of MPC1 and MPC2 and transcriptionally repressed them, leading to increased levels of free NADH in the cytosol and nucleus, thus positively feeding back CtBP1's functions. Consequently, restoring MPC1 and MPC2 in human tumor cells decreases free NADH and inhibits melanoma cell proliferation and migration. CONCLUSIONS Our data indicate that MPC1 and MPC2 are principal mediators that link CtBP1-mediated transcription regulation to NADH production. The discovery of CtBP1 as an NADH regulator in addition to being an NADH sensor shows that CtBP1 is at the center of tumor metabolism and transcription control.


Subject(s)
Alcohol Oxidoreductases/metabolism , DNA-Binding Proteins/metabolism , Melanoma/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Alcohol Oxidoreductases/genetics , Animals , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , Gene Expression , Humans , Melanoma/genetics , Mice , Mitochondrial Membrane Transport Proteins/genetics , Monocarboxylic Acid Transporters , Oxidation-Reduction , Promoter Regions, Genetic , Proprotein Convertase 1/genetics , Proprotein Convertase 1/metabolism , Transcription Factors/metabolism
11.
Diabetologia ; 60(3): 453-463, 2017 03.
Article in English | MEDLINE | ID: mdl-27999871

ABSTRACT

AIMS/HYPOTHESIS: A contributor to beta cell failure in type 2 diabetes and islet transplants is amyloid formation by aggregation of the beta cell peptide, islet amyloid polypeptide (IAPP). Similar to the proinsulin processing pathway that generates insulin, IAPP is derived from a prohormone precursor, proIAPP, which requires cleavage by prohormone convertase (PC) 1/3 and PC2 in rodent pancreatic beta cells. We hypothesised that loss of PC2 would promote beta cell death and dysfunction in a rodent model of human beta cell proIAPP overexpression. METHODS: We generated an islet transplant model wherein immune-deficient mouse models of diabetes received islets expressing amyloidogenic human proIAPP and lacking PC2, leading to restoration of normoglycaemia accompanied by increased secretion of human proIAPP. Blood glucose levels were analysed for up to 16 weeks in transplant recipients and grafts were assessed for islet amyloid and beta cell number and death. RESULTS: Hyperglycaemia (blood glucose >16.9 mmol/l) returned in 94% of recipients of islets expressing human proIAPP and lacking PC2, whereas recipients of islets that express human proIAPP and normal PC2 levels remained normoglycaemic for at least 16 weeks. Islet graft failure was accompanied by a ∼20% reduction in insulin-positive cells, yet the degree of amyloid deposition and beta cell apoptosis was similar to those of controls expressing human proIAPP with functional PC2 levels. CONCLUSIONS/INTERPRETATION: PC2 deficiency in transplanted mouse islets expressing human proIAPP promotes beta cell loss and graft failure. Our data suggest that impaired NH2-terminal processing and increased secretion of human proIAPP promote beta cell failure.


Subject(s)
Amyloid/metabolism , Insulin-Secreting Cells/metabolism , Proprotein Convertase 2/metabolism , Amyloid/genetics , Animals , Blood Glucose/metabolism , Blotting, Western , Humans , Islet Amyloid Polypeptide/metabolism , Islets of Langerhans Transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Proinsulin/metabolism , Proprotein Convertase 1/genetics , Proprotein Convertase 1/metabolism , Proprotein Convertase 2/genetics
12.
J Biol Chem ; 291(13): 6626-40, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26757816

ABSTRACT

The physiological role of the TGR5 receptor in the pancreas is not fully understood. We previously showed that activation of TGR5 in pancreatic ß cells by bile acids induces insulin secretion. Glucagon released from pancreatic α cells and glucagon-like peptide 1 (GLP-1) released from intestinal L cells regulate insulin secretion. Both glucagon and GLP-1 are derived from alternate splicing of a common precursor, proglucagon by PC2 and PC1, respectively. We investigated whether TGR5 activation in pancreatic α cells enhances hyperglycemia-induced PC1 expression thereby releasing GLP-1, which in turn increases ß cell mass and function in a paracrine manner. TGR5 activation augmented a hyperglycemia-induced switch from glucagon to GLP-1 synthesis in human and mouse islet α cells by GS/cAMP/PKA/cAMP-response element-binding protein-dependent activation of PC1. Furthermore, TGR5-induced GLP-1 release from α cells was via an Epac-mediated PKA-independent mechanism. Administration of the TGR5 agonist, INT-777, to db/db mice attenuated the increase in body weight and improved glucose tolerance and insulin sensitivity. INT-777 augmented PC1 expression in α cells and stimulated GLP-1 release from islets of db/db mice compared with control. INT-777 also increased pancreatic ß cell proliferation and insulin synthesis. The effect of TGR5-mediated GLP-1 from α cells on insulin release from islets could be blocked by GLP-1 receptor antagonist. These results suggest that TGR5 activation mediates cross-talk between α and ß cells by switching from glucagon to GLP-1 to restore ß cell mass and function under hyperglycemic conditions. Thus, INT-777-mediated TGR5 activation could be leveraged as a novel way to treat type 2 diabetes mellitus.


Subject(s)
Cholic Acids/pharmacology , Diabetes Mellitus, Experimental/genetics , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Paracrine Communication/genetics , Receptors, G-Protein-Coupled/agonists , Animals , Benzene Derivatives/pharmacology , Benzenesulfonates/pharmacology , Cell Line , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Estrenes/pharmacology , Gene Expression Regulation , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Like Peptide 1/genetics , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Homeostasis/drug effects , Humans , Insulin Resistance , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proprotein Convertase 1/genetics , Proprotein Convertase 1/metabolism , Proprotein Convertase 2/genetics , Proprotein Convertase 2/metabolism , Pyrrolidinones/pharmacology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Sulfones/pharmacology
13.
J Autoimmun ; 80: 48-55, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28238527

ABSTRACT

Despite substantial advances in the research exploring the pathogenesis of Type 1 Diabetes (T1D), the pathophysiological mechanisms involved remain unknown. We hypothesized in this study that interferon alpha (IFNα) participates in the early stages of T1D development by triggering endoplasmic reticulum (ER) stress. To verify our hypothesis, human islets and human EndoC-ßH1 cells were exposed to IFNα and tested for ER stress markers, glucose stimulated insulin secretion (GSIS) and insulin content. IFNα treatment induced upregulation of ER stress markers including Binding immunoglobulin Protein, phospho-eukaryotic translation initiation factor 2α, spliced- X-box binding protein-1, C/EBP homologous protein and activating transcription factor 4. Intriguingly, IFNα treatment did not impair GSIS but significantly decreased insulin production in both human islets and EndoC-ßH1 cells. Furthermore, IFNα decreased the expression of both proinsulin convertase 1 and proinsulin convertase 2, suggesting an altered functional state of the beta cells characterized by a slower proinsulin-insulin conversion. Pretreatment of both human islets and EndoC-ßH1 cells with chemical chaperones 4-phenylbutyric acid and tauroursodeoxycholic acid completely prevented IFNα effects, indicating an ER stress-mediated impairment of insulin production. We demonstrated for the first time that IFNα elicits ER stress in human beta cells providing a novel mechanistic role for this virus-induced cytokine in the development of T1D. Compounds targeting molecular processes altered in ER-stressed beta cells could represent a potential therapeutic strategy to prevent IFNα-induced beta cell dysfunction in the early onset of T1D.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Endoplasmic Reticulum Stress , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Interferon-alpha/metabolism , Apoptosis , Cells, Cultured , Cytokines/metabolism , Humans , Phenylbutyrates/pharmacology , Proprotein Convertase 1/metabolism , Proprotein Convertase 2/metabolism , Taurochenodeoxycholic Acid/pharmacology , Transcription Factor CHOP/metabolism , X-Box Binding Protein 1/metabolism
14.
Bratisl Lek Listy ; 118(10): 609-612, 2017.
Article in English | MEDLINE | ID: mdl-29198128

ABSTRACT

BACKGROUND: Pro-protein processing mechanism plays an important role in neuron injury. OBJECTIVE: To study the protein convertase 1 (PC1) mediated processing mechanism, the ischemic cellular or tissue proPC1/PC1 or proCgA/CgA (pro-chromogranin A) was analyzed. METHODS: NS20Y differentiated cells were stressed by 0-6 h of oxygen and glucose deprivation (OGD) in glucose-free DMEM and an anaerobic jar environment. Ischemic C57BL/J mouse model was established by performing 60-min of middle cerebral artery occlusion (MCAO) operation and subsequent 4 or 24-h reperfusion. The TUNEL, immunochemistry, and Western blot methods were used to detect protein expression in ischemic cells or tissues. RESULTS: The OGD or MCAO stress caused substantial cell death in a dose-dependent manner (p < 0.05 or 0.01). With the increasing OGD dose, proPC1 and PC1 proteins gradually increased (p < 0.05 or 0.01) whereas proCgA and CgA proteins decreased (p < 0.05). In vivo the proPC1 and PC1 expressions presented with a peak at 4-h and then decreased at 24-h reperfusion (p < 0.05 or 0.01). The tissue proCgA and CgA proteins decreased with the increasing reperfusion time (p < 0.05). CONCLUSIONS: The results suggest that the increasing PC1 expression promoted the transformation of proCgA into CgA or smaller peptides, i.e. Pancreastatin or Secretoneurin, and the PC1 mediated processing plays a critical role (Fig. 4, Ref. 15).


Subject(s)
Cell Death , Chromogranin A/metabolism , Infarction, Middle Cerebral Artery/metabolism , Neurons/metabolism , Proprotein Convertase 1/metabolism , Animals , Blotting, Western , Brain Ischemia/metabolism , Cell Differentiation , Cell Hypoxia , Glucose , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Neuropeptides/metabolism , Secretogranin II/metabolism
15.
Int J Obes (Lond) ; 40(6): 973-81, 2016 06.
Article in English | MEDLINE | ID: mdl-26786350

ABSTRACT

BACKGROUND: The proprotein convertase 1/3 (PC1/3), encoded by proprotein convertase subtilisin/kexin type 1 (PCSK1), cleaves and hence activates several orexigenic and anorexigenic proproteins. Congenital inactivation of PCSK1 leads to obesity in human but not in mice. However, a mouse model harboring the hypomorphic mutation N222D is obese. It is not clear why the mouse models differ in phenotype. METHODS: Gene expression analysis was performed with pancreatic islets from Pcsk1(N222D/N222D) mice. Subsequently, biosynthesis, maturation, degradation and activity were studied in islets, pituitary, hypothalamus and cell lines. Coimmunoprecipitation of PC1/3-N222D and human PC1/3 variants associated with obesity with the endoplasmic reticulum (ER) chaperone BiP was studied in cell lines. RESULTS: Gene expression analysis of islets of Pcsk1(N222D/N222D) mice showed enrichment of gene sets related to the proteasome and the unfolded protein response. Steady-state levels of PC1/3-N222D and in particular the carboxy-terminally processed form were strongly reduced in islets, pituitary and hypothalamus. However, impairment of substrate cleavage was tissue dependent. Proinsulin processing was drastically reduced, while processing of proopiomelanocortin (POMC) to adrenocorticotropic hormone (ACTH) in pituitary was only mildly impaired. Growth hormone expression and IGF-1 levels were normal, indicating near-normal processing of hypothalamic proGHRH. PC1/3-N222D binds to BiP and is rapidly degraded by the proteasome. Analysis of human PC1/3 obesity-associated mutations showed increased binding to BiP and prolonged intracellular retention for all investigated mutations, in particular for PC1/3-T175M, PC1/3-G226R and PC1/3-G593R. CONCLUSIONS: This study demonstrates that the hypomorphic mutation in Pcsk1(N222D) mice has an effect on catalytic activity in pancreatic islets, pituitary and hypothalamus. Reduced substrate processing activity in Pcsk1(N222D/N222D) mice is due to enhanced degradation in addition to reduced catalytic activity of the mutant. PC1/3-N222D binds to BiP, suggesting impaired folding and reduced stability. Enhanced BiP binding is also observed in several human obesity-associated PC1/3 variants, suggesting a common mechanism.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/physiology , Endoplasmic Reticulum/metabolism , Mutation , Obesity/genetics , Proprotein Convertase 1/genetics , Animals , Endoplasmic Reticulum Chaperone BiP , Female , Genetic Predisposition to Disease , HEK293 Cells , Heat-Shock Proteins/metabolism , Humans , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice , Obesity/pathology , Polymorphism, Single Nucleotide/genetics , Proinsulin/metabolism , Proprotein Convertase 1/metabolism , Proteasome Endopeptidase Complex/genetics , Unfolded Protein Response
16.
Cell Tissue Res ; 363(2): 555-65, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26298082

ABSTRACT

Successful implantation requires intimate interactions between a competent blastocyst and a receptive uterus. We recently demonstrated that the aberrant activation of opioid signaling by exogenous ligands adversely affects preimplantation embryonic development and subsequent implantation in mice. However, the underlying machinery governing the dynamic homeostasis of the endogenous opioid system in the uterus during early pregnancy remains elusive. We now show that all three major endogenous opioid precursors are spatiotemporally expressed in the uterus during early pregnancy. Moreover, we observe the well-coordinated expression of the synthetic enzyme prohormone convertases 1/3 (PC1/3) at lower levels and of its inhibitor proprotein convertase subtilisin/kexin type 1 inhibitor (Pcsk1n) and the degrading enzyme membrane metallo-endopeptidase (MME) at higher levels in the receptive uterus. Both estrogen and progestin tend to reduce the uterine levels of opioid ligand precursors in the ovariectomized mouse model. This tight regulation of the endogenous opioid system by PC1/3, Pcsk1n and MME has been further confirmed in physiologically related pseudopregnancy and delayed implantation mouse models. The coordinated regulation of opioid precursor biosynthesis and metabolism helps to create appropriate opioid signaling ensuring uterine receptivity for implantation. Thus, endogenous uterine opioid levels are primarily determined by the coordinated expressions of PC1/3, Pcsk1n and MME under the influence of ovarian progestin and estrogen. Our findings raise an additional cautionary note regarding the effects of opioid abuse on early pregnancy events.


Subject(s)
Analgesics, Opioid/metabolism , Embryo Implantation , Enzymes/metabolism , Uterus/enzymology , Animals , Embryo Implantation/drug effects , Embryo Implantation/genetics , Estrogens/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Metalloendopeptidases/metabolism , Mice , Pregnancy , Progestins/pharmacology , Proprotein Convertase 1/metabolism , RNA, Messenger/genetics , Uterus/drug effects
17.
J Immunol ; 193(10): 5118-28, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25297876

ABSTRACT

Nucleic acid-sensing TLRs are involved in both antimicrobial immune responses and autoimmune inflammation. TLR8 is phylogenetically and structurally related to TLR7 and TLR9, which undergo proteolytic processing in the endolysosomes to generate functional receptors. Recent structural analyses of human TLR8 ectodomain and its liganded form demonstrated that TLR8 is also cleaved, and both the N- and C-terminal halves contribute to ligand binding. However, the structures and ssRNA recognition mode of endogenous TLR8 in human primary cells are largely unknown. In this study, we show that proteolytic processing of TLR8 occurs in human monocytes and macrophages in a different manner compared with TLR7/9 cleavage. The insertion loop between leucine-rich repeats 14 and 15 in TLR8 is indispensable for the cleavage and stepwise processing that occurs in the N-terminal fragment. Both furin-like proprotein convertase and cathepsins contribute to TLR8 cleavage in the early/late endosomes. TLR8 recognizes viral ssRNA and endogenous RNA, such as microRNAs, resulting in the production of proinflammatory cytokines. Hence, localization sites of the receptors are crucial for the nucleic acid-sensing mode and downstream signaling.


Subject(s)
Endosomes/metabolism , Macrophages/metabolism , Monocytes/metabolism , RNA, Viral/metabolism , Toll-Like Receptor 8/metabolism , Amino Acid Sequence , Cathepsins/metabolism , Gene Expression , HEK293 Cells , Humans , Ligands , Lysosomes/metabolism , Macrophages/chemistry , Macrophages/cytology , MicroRNAs/chemistry , MicroRNAs/metabolism , Molecular Sequence Data , Monocytes/chemistry , Monocytes/cytology , Primary Cell Culture , Proprotein Convertase 1/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Proteolysis , RNA, Viral/chemistry , Signal Transduction , Toll-Like Receptor 7/chemistry , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/chemistry , Toll-Like Receptor 8/genetics , Toll-Like Receptor 9/chemistry , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism
18.
J Pediatr Gastroenterol Nutr ; 62(4): 577-80, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26488123

ABSTRACT

Autosomal recessive proprotein convertase 1/3 (PC1/3) deficiency, caused by mutations in the PCSK1 gene, is characterized by severe congenital malabsorptive diarrhea, early-onset obesity, and certain endocrine abnormalities. We suspected PC1/3 deficiency in a 4-month-old girl based on the presence of congenital diarrhea and polyuria. Sequencing the whole coding region and splice sites detected a novel homozygous PCSK1 splice-site mutation, c.544-2A>G, in the patient. The mutation resulted in the skipping of exon 5, the generation of a premature termination codon, and nonsense-mediated PCSK1 messenger ribonucleic acid decay, which was demonstrated in complementary DNA derived from fibroblasts.


Subject(s)
Endocrine System Diseases/diagnosis , Mutation , Obesity/diagnosis , Proprotein Convertase 1/deficiency , Cells, Cultured , Codon, Nonsense , DNA Mutational Analysis , Early Diagnosis , Endocrine System Diseases/genetics , Endocrine System Diseases/physiopathology , Endocrine System Diseases/therapy , Female , Growth Disorders/etiology , Growth Disorders/prevention & control , Homozygote , Humans , Infant , Obesity/genetics , Obesity/physiopathology , Obesity/therapy , Parenteral Nutrition , Proprotein Convertase 1/chemistry , Proprotein Convertase 1/genetics , Proprotein Convertase 1/metabolism , RNA Splice Sites , RNA Stability , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Severity of Illness Index , Skin/enzymology , Skin/metabolism , Skin/pathology , Treatment Outcome , Turkey
19.
Hepatology ; 59(6): 2228-37, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24443104

ABSTRACT

UNLABELLED: Fibrolamellar hepatocellular carcinoma (FLC) is a rare subtype of liver cancer occurring mostly in children and young adults. We have shown that FLC comprises two separate entities: pure (p-FLC) and mixed-FLC (m-FLC), differing in clinical presentation and course. We show that p-FLCs have a distinct gene expression signature different from that of m-FLCs, which have a signature similar to that of classical hepatocellular carcinomas. We found p-FLC profiles to be unique among 263 profiles related to diverse tumoral and nontumoral liver samples. We identified two distinct molecular subgroups of p-FLCs with different outcomes. Pathway analysis of p-FLCs revealed ERBB2 overexpression and an up-regulation of glycolysis, possibly leading to compensatory mitochondrial hyperplasia and oncocytic differentiation. Four of the sixteen genes most significantly overexpressed in p-FLCs were neuroendocrine genes: prohormone convertase 1 (PCSK1); neurotensin; delta/notch-like EGF repeat containing; and calcitonin. PCSK1 overexpression was validated by immunohistochemistry, yielding specific, diffuse staining of the protein throughout the cytoplasm, possibly corresponding to a functional form of this convertase. CONCLUSION: p-FLCs have a unique transcriptomic signature characterized by the strong expression of specific neuroendocrine genes, suggesting that these tumors may have a cellular origin different from that of HCC. Our data have implications for the use of genomic profiling for diagnosis and selection of targeted therapies in patients with p-FLC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gene Expression Profiling , Liver Neoplasms/metabolism , Proprotein Convertase 1/metabolism , Receptor, ErbB-2/metabolism , Transcription, Genetic , Adult , Calcitonin/genetics , Calcitonin/metabolism , Female , Genes, erbB-2 , Glycolysis/genetics , Humans , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurotensin/genetics , Neurotensin/metabolism , Proprotein Convertase 1/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
20.
Diabetes Metab Res Rev ; 30(8): 661-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24510483

ABSTRACT

BACKGROUND: Glucagon-like peptide 1 (GLP-1) and glucagon share the same precursor molecule proglucagon, but each arises from a distinct posttranslational process in a tissue-specific manner. Recently, it has been shown that GLP-1 is co-expressed with glucagon in pancreatic islet cells. This study was aimed to investigate the progressive changes of GLP-1 versus glucagon production in pancreatic islets during the course of diabetes development. METHODS: Both type 1 (non-obese diabetes mice) and type 2 (db/db mice) diabetes models were employed in this study. The mice were monitored closely for their diabetes progression and were sacrificed at different stages according to their blood glucose levels. GLP-1 and glucagon expression in the pancreatic islets was examined using immunohistochemistry assays. Quantitative analysis was performed to evaluate the significance of the changes. RESULTS: The ratio of GLP-1-expressing cells to glucagon-expressing cells in the islets showed significant, progressive increase with the development of diabetes in db/db mice. The increase of GLP-1 expression was in agreement with the upregulation of PC1/3 expression in these cells. Interestingly, intra-islet GLP-1 expression was not significantly changed during the development of type 1 diabetes in non-obese diabetes mice. CONCLUSIONS: The study demonstrated that GLP-1 was progressively upregulated in pancreatic islets during type 2 diabetes development. In addition, the data suggest clear differences in intra-islet GLP-1 production between type 1 and type 2 diabetes developments. These differences may have an effect on the clinical and pathophysiological processes of these diseases and may be a target for therapeutic approaches.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide 1/metabolism , Islets of Langerhans/metabolism , Prediabetic State/metabolism , Algorithms , Animals , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Disease Progression , Female , Glucagon/metabolism , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Immunohistochemistry , Islets of Langerhans/pathology , Islets of Langerhans/physiopathology , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Mutant Strains , Prediabetic State/pathology , Prediabetic State/physiopathology , Proprotein Convertase 1/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL