Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 30
1.
Biochem Biophys Rep ; 30: 101250, 2022 Jul.
Article En | MEDLINE | ID: mdl-35295995

The protein wolframin is localized in the membrane of the endoplasmic reticulum (ER), influencing Ca2+ metabolism and ER interaction with mitochondria, but the exact role of the protein remains unclear. Mutations in Wfs1 gene cause autosomal recessive disorder Wolfram syndrome (WS). The first symptom of the WS is diabetes mellitus, so accurate diagnosis of the disease as WS is often delayed. In this study we aimed to characterize the role of the Wfs1 deficiency on bioenergetics of muscles. Alterations in the bioenergetic profiles of Wfs1-exon-5-knock-out (Wfs1KO) male rats in comparison with their wild-type male littermates were investigated using high-resolution respirometry, and enzyme activity measurements. The changes were followed in oxidative (cardiac and soleus) and glycolytic (rectus femoris and gastrocnemius) muscles. There were substrate-dependent alterations in the oxygen consumption rate in Wfs1KO rat muscles. In soleus muscle, decrease in respiration rate was significant in all the followed pathways. The relatively small alterations in muscle during development of WS, such as increased mitochondrial content and/or increase in the OxPhos-related enzymatic activity could be an adaptive response to changes in the metabolic environment. The significant decrease in the OxPhos capacity is substrate dependent indicating metabolic inflexibility when multiple substrates are available.

2.
Cells ; 10(11)2021 11 16.
Article En | MEDLINE | ID: mdl-34831417

Wolfram syndrome (WS), also known as a DIDMOAD (diabetes insipidus, early-onset diabetes mellitus, optic nerve atrophy and deafness) is a rare autosomal disorder caused by mutations in the Wolframin1 (WFS1) gene. Previous studies have revealed that glucagon-like peptide-1 receptor agonist (GLP1 RA) are effective in delaying and restoring blood glucose control in WS animal models and patients. The GLP1 RA liraglutide has also been shown to have neuroprotective properties in aged WS rats. WS is an early-onset, chronic condition. Therefore, early diagnosis and lifelong pharmacological treatment is the best solution to control disease progression. Hence, the aim of this study was to evaluate the efficacy of the long-term liraglutide treatment on the progression of WS symptoms. For this purpose, 2-month-old WS rats were treated with liraglutide up to the age of 18 months and changes in diabetes markers, visual acuity, and hearing sensitivity were monitored over the course of the treatment period. We found that treatment with liraglutide delayed the onset of diabetes and protected against vision loss in a rat model of WS. Therefore, early diagnosis and prophylactic treatment with the liraglutide may also prove to be a promising treatment option for WS patients by increasing the quality of life.


Diabetes Mellitus, Experimental/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Hearing Loss, Sensorineural/drug therapy , Liraglutide/therapeutic use , Nerve Degeneration/drug therapy , Visual Pathways/pathology , Wolfram Syndrome/drug therapy , Animals , C-Peptide/metabolism , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Glucagon-Like Peptide-1 Receptor/metabolism , Hearing Loss, Sensorineural/complications , Liraglutide/pharmacology , Male , Nerve Degeneration/complications , Optic Nerve/drug effects , Optic Nerve/pathology , Optic Nerve/ultrastructure , Phenotype , Rats , Visual Pathways/drug effects , Wolfram Syndrome/complications
3.
Genes (Basel) ; 12(11)2021 10 28.
Article En | MEDLINE | ID: mdl-34828323

Wolfram syndrome (WS) 1 is a rare monogenic neurodegenerative disorder caused by mutations in the gene encoding WFS1. Knowledge of the pathophysiology of WS is incomplete and to date, there is no treatment available. Here, we describe early deviations in the renin-angiotensin-aldosterone system (RAAS) and bradykinin pathway (kallikrein kinin system, KKS) observed in a rat model of WS (Wfs1 KO) and the modulative effect of glucagon-like peptide-1 receptor agonist liraglutide (LIR) and anti-epileptic drug valproate (VPA), which have been proven effective in delaying WS progression in WS animal models. We found that the expression of key receptors of the RAAS and KKS, Agtr2 and Bdkrb1, were drastically downregulated both in vitro and in vivo at an early stage in a rat model of WS. Moreover, in Wfs1, KO serum aldosterone levels were substantially decreased and bradykinin levels increased compared to WT animals. Neither treatment nor their combination affected the gene expression levels seen in the Wfs1 KO animals. However, all the treatments elevated serum aldosterone and decreased bradykinin in the Wfs1 KO rats, as well as increasing angiotensin II levels independent of genotype. Altogether, our results indicate that Wfs1 deficiency might disturb the normal functioning of RAAS and KKS and that LIR and VPA have the ability to modulate these systems.


Calmodulin-Binding Proteins/genetics , Down-Regulation , Membrane Proteins/genetics , Receptor, Angiotensin, Type 2/genetics , Receptor, Bradykinin B1/genetics , Wolfram Syndrome/genetics , Aldosterone/blood , Animals , Cells, Cultured , Disease Models, Animal , Down-Regulation/drug effects , Gene Knockout Techniques , Liraglutide/pharmacology , Liraglutide/therapeutic use , Male , Rats , Renin-Angiotensin System/drug effects , Valproic Acid/pharmacology , Valproic Acid/therapeutic use , Wolfram Syndrome/blood , Wolfram Syndrome/drug therapy
4.
Front Oncol ; 11: 698951, 2021.
Article En | MEDLINE | ID: mdl-34381722

Metabolic plasticity is the ability of the cell to adjust its metabolism to changes in environmental conditions. Increased metabolic plasticity is a defining characteristic of cancer cells, which gives them the advantage of survival and a higher proliferative capacity. Here we review some functional features of metabolic plasticity of colorectal cancer cells (CRC). Metabolic plasticity is characterized by changes in adenine nucleotide transport across the outer mitochondrial membrane. Voltage-dependent anion channel (VDAC) is the main protein involved in the transport of adenine nucleotides, and its regulation is impaired in CRC cells. Apparent affinity for ADP is a functional parameter that characterizes VDAC permeability and provides an integrated assessment of cell metabolic state. VDAC permeability can be adjusted via its interactions with other proteins, such as hexokinase and tubulin. Also, the redox conditions inside a cancer cell may alter VDAC function, resulting in enhanced metabolic plasticity. In addition, a cancer cell shows reprogrammed energy transfer circuits such as adenylate kinase (AK) and creatine kinase (CK) pathway. Knowledge of the mechanism of metabolic plasticity will improve our understanding of colorectal carcinogenesis.

5.
Sci Rep ; 11(1): 2275, 2021 01 26.
Article En | MEDLINE | ID: mdl-33500541

Wolfram syndrome (WS) is a monogenic progressive neurodegenerative disease and is characterized by various neurological symptoms, such as optic nerve atrophy, loss of vision, cognitive decline, memory impairment, and learning difficulties. GLP1 receptor agonist liraglutide and BDNF mimetic 7,8-dihydroxyflavone (7,8-DHF) have had protective effect to visual pathway and to learning and memory in different rat models of neurodegenerative disorders. Although synergistic co-treatment effect has not been reported before and therefore the aim of the current study was to investigate liraglutide, 7,8-DHF and most importantly for the first time their co-treatment effect on degenerative processes in WS rat model. We took 9 months old WS rats and their wild-type (WT) control animals and treated them daily with liraglutide, 7,8-DHF or with the combination of liraglutide and 7,8-DHF up to the age of 12.5 months (n = 47, 5-8 per group). We found that liraglutide, 7,8-DHF and their co-treatment all prevented lateral ventricle enlargement, improved learning in Morris Water maze, reduced neuronal inflammation, delayed the progression of optic nerve atrophy, had remyelinating effect on optic nerve and thereby improved visual acuity in WS rats compared to WT controls. Thus, the use of the liraglutide, 7,8-DHF and their co-treatment could potentially be used as a therapeutic intervention to induce neuroprotection or even neuronal regeneration.


Blindness/drug therapy , Blindness/prevention & control , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/prevention & control , Flavones/therapeutic use , Liraglutide/therapeutic use , Wolfram Syndrome/drug therapy , Animals , Blindness/blood , Blindness/physiopathology , Blood Glucose/metabolism , Body Weight , Calmodulin-Binding Proteins/deficiency , Calmodulin-Binding Proteins/metabolism , Cognitive Dysfunction/blood , Disease Models, Animal , Disease Progression , Drug Therapy, Combination , Fasting/blood , Flavones/pharmacology , Gene Expression Regulation/drug effects , Gene Knockout Techniques , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hyperglycemia/pathology , Learning/drug effects , Liraglutide/pharmacology , Male , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Nerve Degeneration/complications , Nerve Degeneration/pathology , Optic Nerve/drug effects , Optic Nerve/pathology , Optic Nerve/physiopathology , Optic Nerve/ultrastructure , Rats , Remyelination , Visual Acuity/drug effects , Wolfram Syndrome/blood
6.
Sci Rep ; 9(1): 15742, 2019 10 31.
Article En | MEDLINE | ID: mdl-31673100

Wolfram syndrome (WS) is a rare neurodegenerative disorder that is mainly characterized by diabetes mellitus, optic nerve atrophy, deafness, and progressive brainstem degeneration. Treatment with GLP-1 receptor agonists has shown a promising anti-diabetic effect in WS treatment in both animal models and in human patients. Since previous research has tended to focus on investigation of the WS first symptom, diabetes mellitus, the aim of the present study was to examine liraglutide effect on WS-associated neurodegeneration. We took 9-month-old Wfs1 knock-out (KO) animals that already had developed glucose intolerance and treated them with liraglutide for 6 months. Our research results indicate that 6-month liraglutide treatment reduced neuroinflammation and ameliorated endoplasmic reticulum (ER) stress in the inferior olive of the aged WS rat model. Liraglutide treatment also protected retinal ganglion cells from cell death and optic nerve axons from degeneration. According to this, the results of the present study provide novel insight that GLP-1 receptor agonist liraglutide has a neuroprotective effect in the WS rat model.


Glucagon-Like Peptide-1 Receptor/agonists , Liraglutide/therapeutic use , Neuroprotective Agents/therapeutic use , Wolfram Syndrome/drug therapy , Animals , Apoptosis/drug effects , Calmodulin-Binding Proteins/deficiency , Calmodulin-Binding Proteins/genetics , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Glucose Tolerance Test , Hyperglycemia/pathology , Hyperglycemia/prevention & control , Liraglutide/pharmacology , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Neurons/physiology , Neuroprotective Agents/pharmacology , Optic Nerve/metabolism , Rats , Rats, Transgenic , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Wolfram Syndrome/metabolism , Wolfram Syndrome/pathology
7.
Heliyon ; 5(9): e02475, 2019 Sep.
Article En | MEDLINE | ID: mdl-31687570

An intravenously injectable illicit drug made by mixing pseudoephedrine, potassium permanganate, vinegar and water, yielding methcathinone (Mcat) and manganese (Mn), induces an extrapyramidal syndrome with parkinsonism, dystonia, gait and balance disorders similar to manganism. Although the cause of the syndrome is largely attributed to Mn, the interaction of the drug's individual components is not known and the role of Mcat is possibly underestimated. Aim of the present study was to analyze dose-dependent behavioral effects of the mixture and its two main active components Mcat and Mn in an acute setting and determine the lethal doses of each substance. Three groups of C57BL/6 mice were injected intraperitoneally with (1) the drug mixture containing 10, 25, 50, 100 or 150 mg of Mcat and respectively 1.6, 3.8, 6.9, 17.1 and 22.6 mg of Mn per kilogram of body weight; (2) 10, 25, 50, 100, 150, 200 or 300 mg of racemic Mcat/kg of body weight; (3) MnCl2 10, 25 or 50 mg/kg of body weight. Locomotor activity of the animals, various signs and time of death were recorded. Lower doses (10 and 25 mg/kg) of Mcat had a clear motor activity stimulating effect and this was clearly dose-dependent. High doses of Mcat produced epileptic seizures in 74% of the animals and became lethal with the highest doses. Similarly, the mixture had a clear dose-dependent stimulating effect and the higher doses became lethal. The LD50 of the pseudoephedrine mixture was 110.2 mg of Mcat/kg and for pure Mcat 201.7 mg/kg. Mn did not prove to be lethal in doses up to 50 mg/kg, but had a strong dose dependent inhibitory effect on the animals' behavior. Our data reveal that both Mn and Mcat have a significant role in the toxicity of the mixture.

8.
Sci Rep ; 8(1): 10183, 2018 07 05.
Article En | MEDLINE | ID: mdl-29976929

Wolfram syndrome (WS) is a rare autosomal recessive disorder caused by mutations in the WFS1 (Wolframin1) gene. The syndrome first manifests as diabetes mellitus, followed by optic nerve atrophy, deafness, and neurodegeneration. The underlying mechanism is believed to be a dysregulation of endoplasmic reticulum (ER) stress response, which ultimately leads to cellular death. Treatment with glucagon-like peptide-1 (GLP-1) receptor agonists has been shown to normalize ER stress response in several in vitro and in vivo models. Early chronic intervention with the GLP-1 receptor agonist liraglutide starting before the onset of metabolic symptoms prevented the development of glucose intolerance, improved insulin and glucagon secretion control, reduced ER stress and inflammation in Langerhans islets in Wfs1 mutant rats. Thus, treatment with GLP-1 receptor agonists might be a promising strategy as a preventive treatment for human WS patients.


Glucose Intolerance/prevention & control , Incretins/administration & dosage , Liraglutide/administration & dosage , Wolfram Syndrome/complications , Animals , Blood Glucose/analysis , Blood Glucose/metabolism , Calmodulin-Binding Proteins/genetics , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Gene Knockout Techniques , Glucagon/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose Intolerance/blood , Glucose Intolerance/diagnosis , Glucose Intolerance/etiology , Glucose Tolerance Test , Humans , Injections, Subcutaneous , Insulin/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Membrane Proteins/genetics , Rats , Rats, Transgenic , Treatment Outcome , Wolfram Syndrome/genetics
9.
OMICS ; 21(12): 721-732, 2017 12.
Article En | MEDLINE | ID: mdl-29257731

Wolfram syndrome 1 is a rare autosomal recessive neurodegenerative disease characterized by diabetes insipidus, diabetes mellitus, optic atrophy, and deafness. Mutations in the WFS1 gene encoding the wolframin glycoprotein can lead to endoplasmic reticulum stress and unfolded protein responses in cells, but the pathophysiology at whole organism level is poorly understood. In this study, several organs (heart, liver, kidneys, and pancreas) and bodily fluids (trunk blood and urine) of 2- and 6-month old Wfs1 knockout (KO), heterozygote (HZ), and wild-type (WT) mice were analyzed by untargeted and targeted metabolomics using liquid chromatography-mass spectrometry. The key findings were significant perturbations in the metabolism of pancreas and heart before the onset of related clinical signs such as glycosuria that precedes hyperglycemia and thus implies a kidney dysfunction before the onset of classical diabetic nephropathy. The glucose use and gluconeogenesis in KO mice are intensified in early stages, but later the energetic needs are mainly covered by lipolysis. Furthermore, in young mice liver and trunk blood hypouricemia, which in time turns to hyperuricemia, was detected. In summary, we show that the metabolism in Wfs1-deficient mice markedly differs from the metabolism of WT mice in many aspects and discuss the future biological and clinical relevance of these observations.


Membrane Proteins/genetics , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism , Animals , Endoplasmic Reticulum Stress/genetics , Metabolomics/methods , Mice , Mice, Knockout
10.
Mol Med Rep ; 16(5): 7092-7097, 2017 Nov.
Article En | MEDLINE | ID: mdl-28901522

Wolfram syndrome 1 (WS) is a rare neurodegenerative disease that is caused by mutations in the Wolfram syndrome 1 (WFS1) gene, which encodes the endoplasmic reticulum (ER) glycoprotein wolframin. The pathophysiology of WS is ER stress, which is generally considered to induce oxidative stress. As WS has a well­defined monogenetic origin and a model for chronic ER stress, the present study aimed to characterize how glutathione (GSH), a major intracellular antioxidant, was related to the disease and its progression. The concentration of GSH and the activities of reduction/oxidation system enzymes GSH peroxidase and GSH reductase were measured in Wfs1­deficient mice. The GSH content was lower in most of the studied tissues, and the activities of antioxidative enzymes varied between the heart, kidneys and liver tissues. The results indicated that GSH may be needed for ER stress control; however, chronic ER stress from the genetic syndrome eventually depletes the cellular GSH pool and leads to increased oxidative stress.


Glutathione/metabolism , Membrane Proteins/genetics , Wolfram Syndrome/pathology , Aging , Animals , Endoplasmic Reticulum Stress , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Kidney/enzymology , Liver/enzymology , Male , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/enzymology , Pancreas/enzymology , Wolfram Syndrome/genetics
11.
Sci Rep ; 7(1): 10220, 2017 08 31.
Article En | MEDLINE | ID: mdl-28860598

Wolfram syndrome (WS) is a rare autosomal-recessive disorder that is caused by mutations in the WFS1 gene and is characterized by juvenile-onset diabetes, optic atrophy, hearing loss and a number of other complications. Here, we describe the creation and phenotype of Wfs1 mutant rats, in which exon 5 of the Wfs1 gene is deleted, resulting in a loss of 27 amino acids from the WFS1 protein sequence. These Wfs1-ex5-KO232 rats show progressive glucose intolerance, which culminates in the development of diabetes mellitus, glycosuria, hyperglycaemia and severe body weight loss by 12 months of age. Beta cell mass is reduced in older mutant rats, which is accompanied by decreased glucose-stimulated insulin secretion from 3 months of age. Medullary volume is decreased in older Wfs1-ex5-KO232 rats, with the largest decreases at the level of the inferior olive. Finally, older Wfs1-ex5-KO232 rats show retinal gliosis and optic nerve atrophy at 15 months of age. Electron microscopy revealed axonal degeneration and disorganization of the myelin in the optic nerves of older Wfs1-ex5-KO232 rats. The phenotype of Wfs1-ex5-KO232 rats indicates that they have the core symptoms of WS. Therefore, we present a novel rat model of WS.


Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Sequence Deletion , Wolfram Syndrome/genetics , Animals , Disease Models, Animal , Exons , Female , Gliosis/genetics , Gliosis/metabolism , Glucose Intolerance , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Male , Optic Atrophy/genetics , Optic Atrophy/metabolism , Phenotype , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Weight Loss , Wolfram Syndrome/metabolism
12.
PLoS One ; 12(3): e0172825, 2017.
Article En | MEDLINE | ID: mdl-28267787

During amniote evolution, the construction of the forebrain has diverged across different lineages, and accompanying the structural changes, functional diversification of the homologous brain regions has occurred. This can be assessed by studying the expression patterns of marker genes that are relevant in particular functional circuits. In all vertebrates, the dopaminergic system is responsible for the behavioral responses to environmental stimuli. Here we show that the brain regions that receive dopaminergic input through dopamine receptor D1 are relatively conserved, but with some important variations between three evolutionarily distant vertebrate lines-house mouse (Mus musculus), domestic chick (Gallus gallus domesticus) / common quail (Coturnix coturnix) and red-eared slider turtle (Trachemys scripta). Moreover, we find that in almost all instances, those brain regions expressing D1-like dopamine receptor genes also express Wfs1. Wfs1 has been studied primarily in the pancreas, where it regulates the endoplasmic reticulum (ER) stress response, cellular Ca2+ homeostasis, and insulin production and secretion. Using radioligand binding assays in wild type and Wfs1-/- mouse brains, we show that the number of binding sites of D1-like dopamine receptors is increased in the hippocampus of the mutant mice. We propose that the functional link between Wfs1 and D1-like dopamine receptors is evolutionarily conserved and plays an important role in adjusting behavioral reactions to environmental stimuli.


Brain/metabolism , Gene Expression , Membrane Proteins/genetics , Receptors, Dopamine D1/metabolism , Animals , Biomarkers , Chick Embryo , Hippocampus/metabolism , Immunohistochemistry , Membrane Proteins/metabolism , Mice , Mice, Knockout , Protein Binding , Protein Transport , RNA, Messenger/genetics , Receptors, Dopamine D1/genetics , Receptors, Dopamine D5/genetics , Receptors, Dopamine D5/metabolism
13.
Front Mol Neurosci ; 8: 45, 2015.
Article En | MEDLINE | ID: mdl-26379490

BACKGROUND: Mutations in WFS1 gene cause Wolfram syndrome, which is a rare autosomal recessive disorder, characterized by diabetes insipidus, diabetes mellitus, optic nerve atrophy, and deafness. The WFS1 gene product wolframin is located in the endoplasmic reticulum. Mice lacking this gene exhibit disturbances in the processing and secretion of peptides, such as vasopressin and insulin. In the brain, high levels of the wolframin protein have been observed in the hippocampus, amygdala, and limbic structures. The aim of this study was to investigate the effect of Wfs1 knockout (KO) on peptide processing in mouse hippocampus. A peptidomic approach was used to characterize individual peptides in the hippocampus of wild-type and Wfs1 KO mice. RESULTS: We identified 126 peptides in hippocampal extracts and the levels of 10 peptides differed between Wfs1 KO and wild-type mice at P < 0.05. The peptide with the largest alteration was little-LEN, which level was 25 times higher in the hippocampus of Wfs1 KO mice compared to wild-type mice. Processing (cleavage) of little-LEN from the Pcsk1n gene product proSAAS involves prohormone convertase 2 (PC2). Thus, PC2 activity was measured in extracts prepared from the hippocampus of Wfs1 KO mice. The activity of PC2 in Wfs1 mutant mice was significantly higher (149.9 ± 2.3%, p < 0.0001, n = 8) than in wild-type mice (100.0 ± 7.0%, n = 8). However, Western blot analysis showed that protein levels of 7B2, proPC2 and PC2 were same in both groups, and so were gene expression levels. CONCLUSION: Processing of proSAAS is altered in the hippocampus of Wfs1-KO mice, which is caused by increased activity of PC2. Increased activity of PC2 in Wfs1 KO mice is not caused by alteration in the levels of PC2 protein. Our results suggest a functional link between Wfs1 and PC2. Thus, the detailed molecular mechanism of the role of Wfs1 in the regulation of PC2 activity needs further investigation.

14.
PPAR Res ; 2014: 349525, 2014.
Article En | MEDLINE | ID: mdl-24799886

Valproic acid (VPA) is a widely used anticonvulsant and mood-stabilizing drug whose use is often associated with drug-induced weight gain. Treatment with VPA has been shown to upregulate Wfs1 expression in vitro. Aim of the present study was to compare the effect of chronic VPA treatment in wild type (WT) and Wfs1 knockout (KO) mice on hepatic gene expression profile. Wild type, Wfs1 heterozygous, and homozygous mice were treated with VPA for three months (300 mg/kg i.p. daily) and gene expression profiles in liver were evaluated using Affymetrix Mouse GeneChip 1.0 ST array. We identified 42 genes affected by Wfs1 genotype, 10 genes regulated by VPA treatment, and 9 genes whose regulation by VPA was dependent on genotype. Among the genes that were regulated differentially by VPA depending on genotype was peroxisome proliferator-activated receptor delta (Ppard), whose expression was upregulated in response to VPA treatment in WT, but not in Wfs1 KO mice. Thus, regulation of Ppard by VPA is dependent on Wfs1 genotype.

15.
Behav Brain Res ; 244: 90-9, 2013 May 01.
Article En | MEDLINE | ID: mdl-23396150

Immunohistological studies suggest abundant expression of Wfs1 protein in neurons and nerve fibers that lie in the vicinity of dopaminergic (DA-ergic) fibers and neurons. Therefore, we sought to characterize the function of DA-ergic system in Wfs1-deficient mice. In wild-type mice, amphetamine, an indirect agonist of DA, caused significant hyperlocomotion and increase in tissue DA levels in the dorsal and ventral striatum. Both effects of amphetamine were significantly blunted in homozygous Wfs1-deficient mice. Motor stimulation caused by apomorphine, a direct DA receptor agonist, was somewhat stronger in Wfs1-deficient mice compared to their wild-type littermates. However, apomorphine caused a similar reduction in levels of DA metabolites (3,4-dihydroxyphenylacetic acid and homovanillic acid) in the dorsal and ventral striatum in all genotypes. Behavioral sensitization to repeated treatment with amphetamine (2.5 mg/kg) was observed in wild-type, but not in Wfs1-deficient mice. The expression of DA transporter gene (Dat) mRNA was significantly lower in the midbrain of male and female homozygous mice compared to wild-type littermates. Altogether, the blunted effects of amphetamine and the reduced gene expression of DA transporter are probably indicative of an impaired functioning of the DA-ergic system in Wfs1-deficient mice.


Corpus Striatum/physiology , Dopaminergic Neurons/physiology , Membrane Proteins/deficiency , Membrane Proteins/physiology , Amphetamine/pharmacology , Animals , Apomorphine/pharmacology , Central Nervous System Sensitization/physiology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Dopamine Agents/pharmacology , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/drug effects , Female , Gene Expression/drug effects , Male , Mice , Mice, Congenic , Motor Activity/drug effects , Motor Activity/physiology , Receptors, Dopamine D2/metabolism
16.
Physiol Genomics ; 43(24): 1351-8, 2011 Dec 16.
Article En | MEDLINE | ID: mdl-22028430

The Wfs1 gene codes for a protein with unknown function, but deficiency in this protein results in a range of neuropsychiatric and neuroendocrine syndromes. In the present study we aimed to find the functional networks influenced by Wfs1 in the hypothalamus. We performed gene expression profiling (Mouse Gene 1.0 ST Arrays) in Wfs1-deficient mice; 305 genes were differentially expressed with nominal P value<0.01. FDR (false discovery rate)-adjusted P values were significant (0.007) only for two genes: C4b (t=9.66) and Wfs1 (t=-9.03). However, several genes related to G protein signaling were very close to the FDR-adjusted significance level, such as Rgs4 (regulator of G protein signaling 4) that was downregulated (-0.34, t=-5.4) in Wfs1-deficient mice. Changes in Rgs4 and C4b expression were confirmed by QRT-PCR. In humans, Rgs4 is in the locus for bipolar disease (BPD), and its expression is downregulated in BPD. C4b is a gene related to the neurodegenerative diseases. Functional analysis including the entire data set revealed significant alterations in the canonical pathway "G protein-coupled receptor signaling." The gene expression profile in the hypothalami of the Wfs1 mutant mice was significantly similar to the profiles of following biological functions: psychological disorders, bipolar disorder, mood disorder. In conclusion, hypothalamic gene expression profile resembles with some molecular pathways functionally related to the clinical syndromes in the Wolfram syndrome patients.


GTP-Binding Proteins/metabolism , Gene Expression Profiling , Hypothalamus/metabolism , Membrane Proteins/genetics , Signal Transduction/genetics , Animals , Disease/genetics , Gene Expression Regulation , Gene Regulatory Networks/genetics , Guanosine Triphosphate/metabolism , Membrane Proteins/deficiency , Mice , Mice, Knockout , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction
17.
J. physiol. biochem ; 67(3): 381-390, sept. 2011.
Article En | IBECS | ID: ibc-122603

No disponible


Valproic acid (VLP) is a widely used anticonvulsant and mood-stabilizing drug that relieves the endoplasmic reticulum (ER) stress response, a pathogenetic process related to diabetes. The aim of the present study was to evaluate whether acute valproic acid is able to interfere with glucose intolerance in two different diabetes models: The first model was a Wfs1 mutant mouse with an elevated ER stress response and the second model a streptozocin-induced diabetic mouse. VLP (300 mg/kg, i.p.) was administered to Wfs1 knockout (KO) mice and glucose tolerance test was performed 15 min later. VLP did not have an effect on the course of the glucose tolerance test in wild-type mice, while it did normalize the glucose intolerance in Wfs1 knockout mice. Acute valproic acid also lowered the blood glucose levels in streptozocin-treated mice and potentiated the effect of insulin in these mice. Thus, acute valproic acid is effective in lowering blood glucose levels possibly by potentiating insulin action in both Wfs1 KO mice and in streptozocin-induced type 1 diabetic mice (AU)


Animals , Mice , Valproic Acid/pharmacokinetics , Diabetes Mellitus, Experimental/drug therapy , Streptozocin/pharmacokinetics , Hyperglycemia/drug therapy , Disease Models, Animal , Protective Agents/pharmacokinetics
18.
J Physiol Biochem ; 67(3): 381-90, 2011 Sep.
Article En | MEDLINE | ID: mdl-21461749

Valproic acid (VLP) is a widely used anticonvulsant and mood-stabilizing drug that relieves the endoplasmic reticulum (ER) stress response, a pathogenetic process related to diabetes. The aim of the present study was to evaluate whether acute valproic acid is able to interfere with glucose intolerance in two different diabetes models: The first model was a Wfs1 mutant mouse with an elevated ER stress response and the second model a streptozocin-induced diabetic mouse. VLP (300 mg/kg, i.p.) was administered to Wfs1 knockout (KO) mice and glucose tolerance test was performed 15 min later. VLP did not have an effect on the course of the glucose tolerance test in wild-type mice, while it did normalize the glucose intolerance in Wfs1 knockout mice. Acute valproic acid also lowered the blood glucose levels in streptozocin-treated mice and potentiated the effect of insulin in these mice. Thus, acute valproic acid is effective in lowering blood glucose levels possibly by potentiating insulin action in both Wfs1 KO mice and in streptozocin-induced type 1 diabetic mice.


Drug Resistance/genetics , Hypoglycemic Agents/pharmacology , Membrane Proteins/genetics , Mutation , Streptozocin/pharmacology , Valproic Acid/pharmacology , Animals , Blood Glucose/genetics , Body Weight/genetics , Creatinine/urine , Glucose , Glycosuria , Insulin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreas/drug effects , Pancreas/pathology
19.
J. physiol. biochem ; 67(1): 53-60, mar. 2011.
Article En | IBECS | ID: ibc-122634

No disponible


Loss of function of the Wfs1 gene causes Wolfram syndrome, a rare multisystem degenerative disorder. Mutant mice with targeted Wfs1 gene disruption (Wfs1 KO) display morphological and behavioral impairments that are not well understood. The present study aimed to investigate the striatal dopamine output of wild-type, heterozygous, and homozygous Wfs1 null-mutant mice using in vivo microdialysis technique. The baseline dopamine output in striatum was similar in all three animal groups. The application of 100 mM [K+]-rich modified Ringer solution caused in homozygous Wfs1 mutant mice an increase of dopamine output by 400%, while in wild-type and heterozygous animals, the increase of the dopamine output yielded up to 1,200%. In sum, the homozygous Wfs1 mutant (..) (AU)


Animals , Mice , Wolfram Syndrome/physiopathology , Dopamine , Visual Cortex/physiopathology , Potassium/pharmacokinetics , Rats, Inbred WF , Mutation , Conduct Disorder/physiopathology
20.
J Physiol Biochem ; 67(1): 53-60, 2011 Mar.
Article En | MEDLINE | ID: mdl-20972658

Loss of function of the Wfs1 gene causes Wolfram syndrome, a rare multisystem degenerative disorder. Mutant mice with targeted Wfs1 gene disruption (Wfs1 KO) display morphological and behavioral impairments that are not well understood. The present study aimed to investigate the striatal dopamine output of wild-type, heterozygous, and homozygous Wfs1 null-mutant mice using in vivo microdialysis technique. The baseline dopamine output in striatum was similar in all three animal groups. The application of 100 mM [K+]-rich modified Ringer solution caused in homozygous Wfs1 mutant mice an increase of dopamine output by 400%, while in wild-type and heterozygous animals, the increase of the dopamine output yielded up to 1,200%. In sum, the homozygous Wfs1 mutant mice (AUC0₋3 = 0.212 nM/µl h) show significantly decreased striatal dopamine output in response to high-concentration [K+] challenge as compared with wild-type or heterozygous Wfs1 mutant conspecifics (AUC0₋3 = 0.427 and 0.505 nM/µl h, respectively). This could explain at least some of the behavioral alterations in Wfs1 mutant mice.


Corpus Striatum/metabolism , Dopamine/metabolism , Membrane Proteins/genetics , Mutation/genetics , Potassium Chloride/pharmacology , Wolfram Syndrome/genetics , Wolfram Syndrome/metabolism , Animals , Corpus Striatum/drug effects , Heterozygote , Homozygote , Isotonic Solutions/chemistry , Mice , Mice, Transgenic/genetics , Microdialysis/methods , Ringer's Solution
...