Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
2.
Pract Lab Med ; 33: e00304, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36582817

ABSTRACT

Macroenzymes are high-molecular weight forms of enzymes whose presence in human sera can lead to non-pathological, elevated enzyme activities, resulting in further unnecessary clinical evaluation. Precipitation with polyethylene glycol (PEG) is an efficient method for removing macroforms from patient samples and can therefore be used for their identification. Cut-offs (99. Percentiles) for the PEG precipitation activity (%PPA) for eight routine enzyme activities were determined on Abbott's Alinity c, namely: AST (61%), ALT (70%), GGT (41%), LDH (45%), lipase (56%), ALP (17%), CK (36%) and PAMY (45%). Two macroforms (PAMY and CK) were then identified by gel filtration chromatography. We suggest that a %PPA above the enzyme-specific cut-off makes the presence of a macroform possible while a %PPA ≥80%, i.e. markedly above the cut-off, makes it very likely for all enzymes.

3.
J Inherit Metab Dis ; 44(1): 193-214, 2021 01.
Article in English | MEDLINE | ID: mdl-32754920

ABSTRACT

INTRODUCTION: Long-term outcome is postulated to be different in isolated methylmalonic aciduria caused by mutations in the MMAA gene (cblA type) compared with methylmalonyl-CoA mutase deficiency (mut), but case definition was previously difficult. METHOD: Cross-sectional analysis of data from the European Registry and Network for Intoxication type Metabolic Diseases (Chafea no. December 1, 2010). RESULTS: Data from 28 cblA and 95 mut patients in most cases confirmed by mutation analysis (including 4 new mutations for cblA and 19 new mutations for mut). Metabolic crisis is the predominant symptom leading to diagnosis in both groups. Biochemical disturbances during the first crisis were similar in both groups, as well as the age at diagnosis. Z scores of body height and body weight were similar in both groups at birth, but were significantly lower in the mut group at the time of last visit. Glomerular filtration rate was significantly higher in cblA; and as a consequence, chronic renal failure and related complications were significantly less frequent and renal function could be preserved even in older patients. Neurological complications were predominantly found in the mut subgroup. Methylmalonic acidemia (MMA) levels in urine and plasma were significantly lower in cblA. 27/28 cblA patients were reported to be responsive to cobalamin, only 86% of cblA patients were treated with i.m. hydroxocobalamin. In total, 73% of cblA and 98% of mut patients followed a calculated diet with amino acid supplements in 27% (cblA) and 69% (mut). During the study interval, six patients from the mut group died, while all cblA patients survived. CONCLUSION: Although similar at first, cblA patients respond to hydroxocobalamin treatment, subsequently show significantly lower levels of MMA and a milder course than mut patients.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/genetics , Methylmalonyl-CoA Mutase/deficiency , Mitochondrial Membrane Transport Proteins/genetics , Vitamin B 12/metabolism , Amino Acid Metabolism, Inborn Errors/complications , Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/mortality , Child , Cross-Sectional Studies , Female , Glomerular Filtration Rate , Humans , Kidney Failure, Chronic/etiology , Male , Methylmalonic Acid/blood , Methylmalonic Acid/urine , Methylmalonyl-CoA Mutase/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mutation
4.
PLoS One ; 10(6): e0129226, 2015.
Article in English | MEDLINE | ID: mdl-26075596

ABSTRACT

Glucagon regulates glucose homeostasis by controlling glycogenolysis and gluconeogenesis in the liver. Exaggerated and dysregulated glucagon secretion can exacerbate hyperglycemia contributing to type 2 diabetes (T2D). Thus, it is important to understand how glucagon receptor (GCGR) activity and signaling is controlled in hepatocytes. To better understand this, we sought to identify proteins that interact with the GCGR to affect ligand-dependent receptor activation. A Flag-tagged human GCGR was recombinantly expressed in Chinese hamster ovary (CHO) cells, and GCGR complexes were isolated by affinity purification (AP). Complexes were then analyzed by mass spectrometry (MS), and protein-GCGR interactions were validated by co-immunoprecipitation (Co-IP) and Western blot. This was followed by studies in primary hepatocytes to assess the effects of each interactor on glucagon-dependent glucose production and intracellular cAMP accumulation, and then in immortalized CHO and liver cell lines to further examine cell signaling. Thirty-three unique interactors were identified from the AP-MS screening of GCGR expressing CHO cells in both glucagon liganded and unliganded states. These studies revealed a particularly robust interaction between GCGR and 5 proteins, further validated by Co-IP, Western blot and qPCR. Overexpression of selected interactors in mouse hepatocytes indicated that two interactors, LDLR and TMED2, significantly enhanced glucagon-stimulated glucose production, while YWHAB inhibited glucose production. This was mirrored with glucagon-stimulated cAMP production, with LDLR and TMED2 enhancing and YWHAB inhibiting cAMP accumulation. To further link these interactors to glucose production, key gluconeogenic genes were assessed. Both LDLR and TMED2 stimulated while YWHAB inhibited PEPCK and G6Pase gene expression. In the present study, we have probed the GCGR interactome and found three novel GCGR interactors that control glucagon-stimulated glucose production by modulating cAMP accumulation and genes that control gluconeogenesis. These interactors may be useful targets to control glucose homeostasis in T2D.


Subject(s)
Liver/metabolism , Protein Interaction Mapping , Protein Interaction Maps , Proteomics , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Animals , CHO Cells , Carrier Proteins , Cell Line , Cricetulus , Cyclic AMP/metabolism , Gene Expression Regulation , Gluconeogenesis/genetics , Glucose/metabolism , Hepatocytes/metabolism , Mice , Protein Binding , Proteomics/methods , Receptors, G-Protein-Coupled , Reproducibility of Results
5.
PLoS One ; 10(3): e0119136, 2015.
Article in English | MEDLINE | ID: mdl-25806541

ABSTRACT

Zinc has an important role in normal pancreatic beta cell physiology as it regulates gene transcription, insulin crystallization and secretion, and cell survival. Nevertheless, little is known about how zinc is transported through the plasma membrane of beta cells and which of the class of zinc influx transporters (Zip) is involved. Zip4 was previously shown to be expressed in human and mouse beta cells; however, its function there is still unknown. Therefore, the aim of this study was to define the zinc transport role of Zip4 in beta cells. To investigate this, Zip4 was over-expressed in MIN6 beta cells using a pCMV6-Zip4GFP plasmid. Organelle staining combined with confocal microscopy showed that Zip4 exhibits a widespread localization in MIN6 cells. Time-lapse zinc imaging experiments showed that Zip4 increases cytoplasmic zinc levels. This resulted in increased granular zinc content and glucose-stimulated insulin secretion. Interestingly, it is unlikely that the increased glucose stimulated insulin secretion was triggered by a modulation of mitochondrial function, as mitochondrial membrane potential remained unchanged. To define the role of Zip4 in-vivo, we generated a beta cell-specific knockout mouse model (Zip4BKO). Deletion of the Zip4 gene was confirmed in Zip4BKO islets by PCR, RT-PCR, and immuno-histochemistry. Zip4BKO mice showed slightly improved glucose homeostasis but no change in insulin secretion during an oral glucose tolerance test. While Zip4 was not found to be essential for proper glucose homeostasis and insulin secretion in vivo in mice, this study also found that Zip4 mediates increases in cytoplasmic and granular zinc pools and stimulates glucose dependant insulin secretion in-vitro.


Subject(s)
Cation Transport Proteins/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Zinc/metabolism , Animals , Biological Transport , Cation Transport Proteins/genetics , Cell Line , Glucose/metabolism , Homeostasis/physiology , Insulin Secretion , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Knockout
6.
Mol Cell Proteomics ; 13(11): 3049-62, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25044020

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is an incretin hormone that regulates glucose homeostasis. Because of their direct stimulation of insulin secretion from pancreatic ß cells, GLP-1 receptor (GLP-1R) agonists are now important therapeutic options for the treatment of type 2 diabetes. To better understand the mechanisms that control the insulinotropic actions of GLP-1, affinity purification and mass spectrometry (AP-MS) were employed to uncover potential proteins that functionally interact with the GLP-1R. AP-MS performed on Chinese hamster ovary cells or MIN6 ß cells, both expressing the human GLP-1R, revealed 99 proteins potentially associated with the GLP-1R. Three novel GLP-1R interactors (PGRMC1, Rab5b, and Rab5c) were further validated through co-immunoprecipitation/immunoblotting, fluorescence resonance energy transfer, and immunofluorescence. Functional studies revealed that overexpression of PGRMC1, a novel cell surface receptor that associated with liganded GLP-1R, enhanced GLP-1-induced insulin secretion (GIIS) with the most robust effect. Knockdown of PGRMC1 in ß cells decreased GIIS, indicative of positive interaction with GLP-1R. To gain insight mechanistically, we demonstrated that the cell surface PGRMC1 ligand P4-BSA increased GIIS, whereas its antagonist AG-205 decreased GIIS. It was then found that PGRMC1 increased GLP-1-induced cAMP accumulation. PGRMC1 activation and GIIS induced by P4-BSA could be blocked by inhibition of adenylyl cyclase/EPAC signaling or the EGF receptor-PI3K signal transduction pathway. These data reveal a dual mechanism for PGRMC1-increased GIIS mediated through cAMP and EGF receptor signaling. In conclusion, we identified several novel GLP-1R interacting proteins. PGRMC1 expressed on the cell surface of ß cells was shown to interact with the activated GLP-1R to enhance the insulinotropic actions of GLP-1.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Membrane Proteins/metabolism , Receptors, Glucagon/metabolism , Receptors, Progesterone/metabolism , Adenylyl Cyclase Inhibitors , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Cyclic AMP/biosynthesis , Cyclic AMP/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Glucagon-Like Peptide-1 Receptor , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/metabolism , Humans , Insulin Secretion , Mass Spectrometry , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Rats , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/genetics , rab5 GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...