Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 24(1)2023 Jan 03.
Article in English | MEDLINE | ID: mdl-36614300

ABSTRACT

Type 2 diabetes (T2D) represents a multifactorial metabolic disease with a strong genetic predisposition. Despite elaborate efforts in identifying the genetic variants determining individual susceptibility towards T2D, the majority of genetic factors driving disease development remain poorly understood. With the aim to identify novel T2D risk genes we previously generated an N2 outcross population using the two inbred mouse strains New Zealand obese (NZO) and C3HeB/FeJ (C3H). A linkage study performed in this population led to the identification of the novel T2D-associated quantitative trait locus (QTL) Nbg15 (NZO blood glucose on chromosome 15, Logarithm of odds (LOD) 6.6). In this study we used a combined approach of positional cloning, gene expression analyses and in silico predictions of DNA polymorphism on gene/protein function to dissect the genetic variants linking Nbg15 to the development of T2D. Moreover, we have generated congenic strains that associated the distal sublocus of Nbg15 to mechanisms altering pancreatic beta cell function. In this sublocus, Cbx6, Fam135b and Kdelr3 were nominated as potential causative genes associated with the Nbg15 driven effects. Moreover, a putative mutation in the Kdelr3 gene from NZO was identified, negatively influencing adaptive responses associated with pancreatic beta cell death and induction of endoplasmic reticulum stress. Importantly, knockdown of Kdelr3 in cultured Min6 beta cells altered insulin granules maturation and pro-insulin levels, pointing towards a crucial role of this gene in islets function and T2D susceptibility.


Subject(s)
Diabetes Mellitus, Type 2 , Genetic Predisposition to Disease , Obesity , Receptors, Peptide , Animals , Mice , Diabetes Mellitus, Type 2/genetics , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Mice, Inbred C3H , Mice, Obese , Obesity/genetics , Receptors, Peptide/genetics
2.
Sci Rep ; 12(1): 14255, 2022 08 22.
Article in English | MEDLINE | ID: mdl-35995803

ABSTRACT

Cystic fibrosis is an autosomal recessive disorder caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) that can lead to terminal respiratory failure. Ultrafine carbonaceous particles, which are ubiquitous in ambient urban and indoor air, are increasingly considered as major contributors to the global health burden of air pollution. However, their effects on the expression of CFTR and associated genes in lung epithelial cells have not yet been investigated. We therefore evaluated the effects of carbon nanoparticles (CNP), generated by spark-ablation, on the human bronchial epithelial cell line 16HBE14o- at air-liquid interface (ALI) culture conditions. The ALI-cultured cells exhibited epithelial barrier integrity and increased CFTR expression. Following a 4-h exposure to CNP, the cells exhibited a decreased barrier integrity, as well as decreased expression of CFTR transcript and protein levels. Furthermore, transcriptomic analysis revealed that the CNP-exposed cells showed signs of oxidative stress, apoptosis and DNA damage. In conclusion, this study describes spark-ablated carbon nanoparticles in a realistic exposure of aerosols to decrease CFTR expression accompanied by transcriptomic signs of oxidative stress, apoptosis and DNA damage.


Subject(s)
Cystic Fibrosis , Nanoparticles , Bronchi/metabolism , Carbon/metabolism , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/metabolism , Humans , Nanoparticles/toxicity , Particulate Matter/metabolism
3.
Diabetes ; 70(12): 2796-2809, 2021 12.
Article in English | MEDLINE | ID: mdl-34561225

ABSTRACT

The two closely related RabGTPase-activating proteins (RabGAPs) TBC1D1 and TBC1D4, both substrates for AMPK, play important roles in exercise metabolism and contraction-dependent translocation of GLUT4 in skeletal muscle. However, the specific contribution of each RabGAP in contraction signaling is mostly unknown. In this study, we investigated the cooperative AMPK-RabGAP signaling axis in the metabolic response to exercise/contraction using a novel mouse model deficient in active skeletal muscle AMPK combined with knockout of either Tbc1d1, Tbc1d4, or both RabGAPs. AMPK deficiency in muscle reduced treadmill exercise performance. Additional deletion of Tbc1d1 but not Tbc1d4 resulted in a further decrease in exercise capacity. In oxidative soleus muscle, AMPK deficiency reduced contraction-mediated glucose uptake, and deletion of each or both RabGAPs had no further effect. In contrast, in glycolytic extensor digitorum longus muscle, AMPK deficiency reduced contraction-stimulated glucose uptake, and deletion of Tbc1d1, but not Tbc1d4, led to a further decrease. Importantly, skeletal muscle deficient in AMPK and both RabGAPs still exhibited residual contraction-mediated glucose uptake, which was completely abolished by inhibition of the GTPase Rac1. Our results demonstrate a novel mechanistic link between glucose transport and the GTPase signaling framework in skeletal muscle in response to contraction.


Subject(s)
Glucose/metabolism , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Biological Transport/genetics , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/genetics , Neuropeptides/metabolism , Physical Conditioning, Animal/physiology , Signal Transduction/genetics , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
4.
Mamm Genome ; 32(3): 153-172, 2021 06.
Article in English | MEDLINE | ID: mdl-33880624

ABSTRACT

Type 2 diabetes (T2D) has a strong genetic component. Most of the gene variants driving the pathogenesis of T2D seem to target pancreatic ß-cell function. To identify novel gene variants acting at early stage of the disease, we analyzed whole transcriptome data to identify differential expression (DE) and alternative exon splicing (AS) transcripts in pancreatic islets collected from two metabolically diverse mouse strains at 6 weeks of age after three weeks of high-fat-diet intervention. Our analysis revealed 1218 DE and 436 AS genes in islets from NZO/Hl vs C3HeB/FeJ. Whereas some of the revealed genes present well-established markers for ß-cell failure, such as Cd36 or Aldh1a3, we identified numerous DE/AS genes that have not been described in context with ß-cell function before. The gene Lgals2, previously associated with human T2D development, was DE as well as AS and localizes in a quantitative trait locus (QTL) for blood glucose on Chr.15 that we reported recently in our N2(NZOxC3H) population. In addition, pathway enrichment analysis of DE and AS genes showed an overlap of only half of the revealed pathways, indicating that DE and AS in large parts influence different pathways in T2D development. PPARG and adipogenesis pathways, two well-established metabolic pathways, were overrepresented for both DE and AS genes, probably as an adaptive mechanism to cope for increased cellular stress. Our results provide guidance for the identification of novel T2D candidate genes and demonstrate the presence of numerous AS transcripts possibly involved in islet function and maintenance of glucose homeostasis.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Galectin 2/genetics , Insulin/genetics , PPAR gamma/genetics , Adipogenesis/genetics , Alternative Splicing/genetics , Animals , Blood Glucose/genetics , CD36 Antigens/genetics , Diabetes Mellitus, Type 2/pathology , Exons/genetics , Gene Expression Regulation/genetics , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/growth & development , Islets of Langerhans/pathology , Metabolic Networks and Pathways/genetics , Mice , Quantitative Trait Loci/genetics , Retinal Dehydrogenase/genetics , Transcriptome/genetics
5.
Genetics ; 210(4): 1527-1542, 2018 12.
Article in English | MEDLINE | ID: mdl-30341086

ABSTRACT

To identify novel disease genes for type 2 diabetes (T2D) we generated two backcross populations of obese and diabetes-susceptible New Zealand Obese (NZO/HI) mice with the two lean mouse strains 129P2/OlaHsd and C3HeB/FeJ. Subsequent whole-genome linkage scans revealed 30 novel quantitative trait loci (QTL) for T2D-associated traits. The strongest association with blood glucose [12 cM, logarithm of the odds (LOD) 13.3] and plasma insulin (17 cM, LOD 4.8) was detected on proximal chromosome 7 (designated Nbg7p, NZO blood glucose on proximal chromosome 7) exclusively in the NZOxC3H crossbreeding, suggesting that the causal gene is contributed by the C3H genome. Introgression of the critical C3H fragment into the genetic NZO background by generating recombinant congenic strains and metabolic phenotyping validated the phenotype. For the detection of candidate genes in the critical region (30-46 Mb), we used a combined approach of haplotype and gene expression analysis to search for C3H-specific gene variants in the pancreatic islets, which appeared to be the most likely target tissue for the QTL. Two genes, Atp4a and Pop4, fulfilled the criteria from our candidate gene approaches. The knockdown of both genes in MIN6 cells led to decreased glucose-stimulated insulin secretion, indicating a regulatory role of both genes in insulin secretion, thereby possibly contributing to the phenotype linked to Nbg7p In conclusion, our combined- and comparative-cross analysis approach has successfully led to the identification of two novel diabetes susceptibility candidate genes, and thus has been proven to be a valuable tool for the discovery of novel disease genes.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Insulin Secretion/genetics , Insulin/genetics , Obesity/genetics , Animals , Blood Glucose/genetics , Chromosome Mapping , Diabetes Mellitus, Type 2/pathology , Genomics , Genotype , Glucose , H(+)-K(+)-Exchanging ATPase/genetics , Humans , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Mice , Mice, Inbred C3H , Mice, Inbred Strains , Obesity/pathology , Quantitative Trait Loci/genetics , Ribonucleases/genetics , Ribonucleoproteins/genetics
6.
Endocrinology ; 159(4): 1748-1761, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29481597

ABSTRACT

The Rab guanosine triphosphatase-activating protein (RabGAP) TBC1D1 has been shown to be a key regulator of glucose and lipid metabolism in skeletal muscle. Its function in pancreatic islets, however, is not yet fully understood. Here, we aimed to clarify the specific impact of TBC1D1 on insulin secretion and substrate use in pancreatic islets. We analyzed the dynamics of glucose-stimulated insulin secretion (GSIS) and lipid metabolism in isolated islets from Tbc1d1-deficient (D1KO) mice. To further investigate the underlying cellular mechanisms, we conducted pharmacological studies in these islets. In addition, we determined morphology and number of both pancreatic islets and insulin vesicles in ß-cells using light and transmission electron microscopy. Isolated pancreatic islets from D1KO mice exhibited substantially increased GSIS compared with wild-type (WT) controls. This was attributed to both enhanced first and second phase of insulin secretion, and this enhanced secretion persisted during repetitive glucose stimuli. Studies with sulfonylureas or KCl in isolated islets demonstrated that TBC1D1 exerts its function via a signaling pathway at the level of membrane depolarization. In line, ultrastructural analysis of isolated pancreatic islets revealed both higher insulin-granule density and number of docked granules in ß-cells from D1KO mice compared with WT controls. Like in skeletal muscle, lipid use in isolated islets was enhanced upon D1KO, presumably as a result of a higher mitochondrial fission rate and/or higher mitochondrial activity. Our results clearly demonstrate a dual role of TBC1D1 in controlling substrate metabolism of the pancreatic islet.


Subject(s)
Fatty Acids/metabolism , GTPase-Activating Proteins/metabolism , Insulin/metabolism , Islets of Langerhans/physiology , Lipid Metabolism/genetics , Animals , GTPase-Activating Proteins/genetics , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Male , Mice , Mice, Knockout
7.
Neuropharmacology ; 107: 100-110, 2016 08.
Article in English | MEDLINE | ID: mdl-27012889

ABSTRACT

Nicotinic acetylcholine receptor (nAChR) subtypes containing the α4 subunit, particularly α4ß2 nAChRs, play an important role in cognitive functioning. The impact of the smoking cessation aid varenicline, a selective partial α4ß2 nAChR agonist, on (1) changes of central protein and mRNA expression of this receptor and (2) on memory deficits in a mouse model of cognitive impairment was investigated. Protein and mRNA expression of both the α4 and ß2 receptor subunits in mouse brain endothelial and hippocampal cells as well as hippocampus and neocortex tissues were determined by western blot and realtime PCR, respectively. The ß2 antibody showed low specificity, though. Tissues were examined following a 2-week oral treatment with various doses of varenicline (0.01, 0.1, 1, 3 mg/kg/day) or vehicle. In addition, episodic memory of mice was assessed following this treatment with an object recognition task using (1) normal mice and (2) animals with anticholinergic-induced memory impairment (i.p. injection of 0.5 mg/kg scopolamine). Varenicline dose-dependently increased protein expression of both the α4 and ß2 subunit in cell cultures and brain tissues, respectively, but had no effect on mRNA expression of both subunits. Scopolamine injection induced a significant reduction of object memory in vehicle-treated mice. By contrast, cognitive performance was not altered by scopolamine in varenicline-treated mice. In conclusion, a 2-week oral treatment with varenicline prevented memory impairment in the scopolamine mouse model. In parallel, protein, but not mRNA expression was upregulated, suggesting a posttranscriptional mechanism. Our findings suggest a beneficial effect of varenicline on cognitive dysfunction.


Subject(s)
Brain/drug effects , Cognition/drug effects , Nootropic Agents/pharmacology , Receptors, Nicotinic/metabolism , Varenicline/pharmacology , Administration, Oral , Animals , Brain/metabolism , Cell Line , Cognition/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Male , Memory Disorders/drug therapy , Memory Disorders/metabolism , Mice, Inbred C57BL , RNA, Messenger/metabolism , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Scopolamine
9.
Diabetes ; 64(3): 746-59, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25249576

ABSTRACT

The Rab-GTPase­activating proteins TBC1D1 and TBC1D4 (AS160) were previously shown to regulate GLUT4 translocation in response to activation of AKT and AMP-dependent kinase [corrected]. However, knockout mice lacking either Tbc1d1 or Tbc1d4 displayed only partially impaired insulin-stimulated glucose uptake in fat and muscle tissue. The aim of this study was to determine the impact of the combined inactivation of Tbc1d1 and Tbc1d4 on glucose metabolism in double-deficient (D1/4KO) mice. D1/4KO mice displayed normal fasting glucose concentrations but had reduced tolerance to intraperitoneally administered glucose, insulin, and AICAR. D1/4KO mice showed reduced respiratory quotient, indicating increased use of lipids as fuel. These mice also consistently showed elevated fatty acid oxidation in isolated skeletal muscle, whereas insulin-stimulated glucose uptake in muscle and adipose cells was almost completely abolished. In skeletal muscle and white adipose tissue, the abundance of GLUT4 protein, but not GLUT4 mRNA, was substantially reduced. Cell surface labeling of GLUTs indicated that RabGAP deficiency impairs retention of GLUT4 in intracellular vesicles in the basal state. Our results show that TBC1D1 and TBC1D4 together play essential roles in insulin-stimulated glucose uptake and substrate preference in skeletal muscle and adipose cells.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Biological Transport/drug effects , GTPase-Activating Proteins/metabolism , Glucose/metabolism , Insulin/pharmacology , Ribonucleotides/pharmacology , Aminoimidazole Carboxamide/pharmacology , Animals , Body Composition/physiology , Body Weight/physiology , Calorimetry, Indirect , GTPase-Activating Proteins/genetics , Genotyping Techniques , Glucose Transporter Type 4/metabolism , Male , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...