Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 51
1.
Gene ; 914: 148313, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38447681

Adenosine kinase deficiency (OMIM #614300) is a type of inborn errors of metabolism with multiorgan symptoms primarily neurological disorders, hepatic impairment, global developmental delay, and mild dysmorphism. The genetic causes of adenosine kinase deficiency are homozygous or compound heterozygous loss-of-function variants of ADK. To date, fewer than 25 cases of adenosine kinase deficiency have been reported worldwide and none have been reported in China. In this research, trio whole-exome sequencing (Trio-WES) identified a novel homozygous ADK (NM_001123.4) out-of-frame deletion, c.518_519delCA (p.Thr173Serfs*15), in a Chinese patient with rare phenotypes of sepsis, metabolites disruption and neutrophil dysfunction. This variant was dysfunctional, with marked reduction of ADK level in both the patient's peripheral blood and cells transfected with the corresponding variant. Additionally, metabolomics detected by high-throughput mass spectrometry showed disturbances in the methionine (Met) and energy pathway. RNA sequencing (RNA-seq) of the patient's peripheral blood suggested a defective anti-inflammatory response characterized by impaired neutrophil activation, migration, and degranulation, which might be the primary cause for the sepsis. To our knowledge, we identified the first Chinese patient of adenosine kinase deficiency with a novel homozygous out-of-frame deletion in ADK causing multiorgan disorders, metabolites disruption, rare phenotypes of sepsis, and neutrophil dysfunction. Our findings broaden the genetic spectrum and pathogenic mechanisms of adenosine kinase deficiency.


Adenosine Kinase , Homozygote , Neutrophils , Phenotype , Sepsis , Humans , Sepsis/genetics , Neutrophils/metabolism , Adenosine Kinase/genetics , Adenosine Kinase/deficiency , Male , Exome Sequencing , Sequence Deletion , Female
2.
Mol Genet Metab ; 132(1): 38-43, 2021 01.
Article En | MEDLINE | ID: mdl-33309011

Adenosine kinase (ADK) deficiency is characterized by liver disease, dysmorphic features, epilepsy and developmental delay. This defect disrupts the adenosine/AMP futile cycle and interferes with the upstream methionine cycle. We report the clinical, histological and biochemical courses of three ADK children carrying two new mutations and presenting with neonatal cholestasis and neurological disorders. One of them died of liver failure whereas the other two recovered from their liver damage. As the phenotype was consistent with a mitochondrial disorder, we studied liver mitochondrial respiratory chain activities in two patients and revealed a combined defect of several complexes. In addition, we retrospectively analyzed methionine plasma concentration, a hallmark of ADK deficiency, in a cohort of children and showed that methionine level in patients with ADK deficiency was strongly increased compared with patients with other liver diseases. ADK deficiency is a cause of neonatal or early infantile liver disease that may mimic primary mitochondrial disorders. In this context, an elevation of methionine plasma levels over twice the upper limit should not be considered as a nonspecific finding. ADK deficiency induced-liver dysfunction is most often transient, but could be life-threatening.


Adenosine Kinase/genetics , Amino Acid Metabolism, Inborn Errors/genetics , Developmental Disabilities/genetics , Epilepsy/genetics , Glycine N-Methyltransferase/deficiency , Adenosine/genetics , Adenosine/metabolism , Adenosine Kinase/deficiency , Amino Acid Metabolism, Inborn Errors/complications , Amino Acid Metabolism, Inborn Errors/pathology , Child , Developmental Disabilities/complications , Developmental Disabilities/pathology , Epilepsy/complications , Epilepsy/pathology , Female , Genetic Predisposition to Disease , Glycine N-Methyltransferase/genetics , Humans , Infant , Infant, Newborn , Liver Diseases/complications , Liver Diseases/genetics , Liver Diseases/pathology , Male , Retrospective Studies
3.
J Endocrinol ; 242(2): 159-172, 2019 08.
Article En | MEDLINE | ID: mdl-31189131

Insulin resistance-related disorders are associated with endothelial dysfunction. Accumulating evidence has suggested a role for adenosine signaling in the regulation of endothelial function. Here, we identified a crucial role of endothelial adenosine kinase (ADK) in the regulation of insulin resistance. Feeding mice with a high-fat diet (HFD) markedly enhanced the expression of endothelial Adk. Ablation of endothelial Adk in HFD-fed mice improved glucose tolerance and insulin sensitivity and decreased hepatic steatosis, adipose inflammation and adiposity, which were associated with improved arteriole vasodilation, decreased inflammation and increased adipose angiogenesis. Mechanistically, ADK inhibition or knockdown in human umbilical vein endothelial cells (HUVECs) elevated intracellular adenosine level and increased endothelial nitric oxide synthase (NOS3) activity, resulting in an increase in nitric oxide (NO) production. Antagonism of adenosine receptor A2b abolished ADK-knockdown-enhanced NOS3 expression in HUVECs. Additionally, increased phosphorylation of NOS3 in ADK-knockdown HUVECs was regulated by an adenosine receptor-independent mechanism. These data suggest that Adk-deficiency-elevated intracellular adenosine in endothelial cells ameliorates diet-induced insulin resistance and metabolic disorders, and this is associated with an enhancement of NO production caused by increased NOS3 expression and activation. Therefore, ADK is a potential target for the prevention and treatment of metabolic disorders associated with insulin resistance.


Adenosine Kinase/deficiency , Endothelium, Vascular/metabolism , Insulin Resistance/physiology , Adenosine Kinase/genetics , Adipose Tissue/metabolism , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Endothelium, Vascular/cytology , Fatty Liver/etiology , Fatty Liver/genetics , Fatty Liver/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/etiology , Inflammation/genetics , Inflammation/metabolism , Insulin Resistance/genetics , Male , Mice, Knockout , Mice, Transgenic , Nitric Oxide Synthase Type III/metabolism , Obesity/etiology , Obesity/genetics , Obesity/metabolism , Phosphorylation
4.
Neuropediatrics ; 50(1): 46-50, 2019 02.
Article En | MEDLINE | ID: mdl-30477030

Adenosine kinase (ADK) deficiency (OMIM [online mendelian inheritance in man]: 614300) is an autosomal recessive disorder of adenosine and methionine metabolism, with a unique clinical phenotype, mainly involving the central nervous system and dysmorphic features. Patients usually present early in life with sepsis-like symptoms, respiratory difficulties, and neonatal jaundice. Subsequently, patients demonstrate hypotonia and global developmental delay. Biochemically, methionine is elevated with normal homocysteine levels and the diagnosis is confirmed through molecular analysis of the ADK gene. There is no curative treatment; however, a methionine-restricted diet has been tried with variable outcomes. Herein, we report a 4-year-old Saudi female with global developmental delay, hypotonia, and dysmorphic features. Interestingly, she has a tall stature, developmental dysplasia of the hip, optic nerve gliosis, and tigroid fundus. We found a mutation not reported previously and we compared the current case with previously reported cases. We alert clinicians to consider ADK deficiency in any neonate presenting with global developmental delay, hypotonia, dysmorphic features, and high methionine levels.


Adenosine Kinase/deficiency , Developmental Disabilities/diagnostic imaging , Developmental Disabilities/enzymology , Muscle Hypotonia/diagnostic imaging , Muscle Hypotonia/enzymology , Child, Preschool , Female , Humans
5.
Arterioscler Thromb Vasc Biol ; 38(12): 2780-2792, 2018 12.
Article En | MEDLINE | ID: mdl-30571174

Objective- Monocyte-derived foam cells are one of the key players in the formation of atherosclerotic plaques. Adenosine receptors and extracellular adenosine have been demonstrated to modulate foam cell formation. ADK (adenosine kinase) is a major enzyme regulating intracellular adenosine levels, but its functional role in myeloid cells remains poorly understood. To enhance intracellular adenosine levels in myeloid cells, ADK was selectively deleted in novel transgenic mice using Cre-LoxP technology, and foam cell formation and the development of atherosclerotic lesions were determined. Approach and Results- ADK was upregulated in macrophages on ox-LDL (oxidized low-density lipoprotein) treatment in vitro and was highly expressed in foam cells in atherosclerotic plaques. Atherosclerotic mice deficient in ADK in myeloid cells were generated by breeding floxed ADK (ADKF/F) mice with LysM-Cre (myeloid-specific Cre recombinase expressing) mice and ApoE-/- (apolipoprotein E deficient) mice. Mice absent ADK in myeloid cells exhibited much smaller atherosclerotic plaques compared with controls. In vitro assays showed that ADK deletion or inhibition resulted in increased intracellular adenosine and reduced DNA methylation of the ABCG1 (ATP-binding cassette transporter G1) gene. Loss of methylation was associated with ABCG1 upregulation, enhanced cholesterol efflux, and eventually decreased foam cell formation. Conclusions- Augmentation of intracellular adenosine levels through ADK knockout in myeloid cells protects ApoE-/- mice against atherosclerosis by reducing foam cell formation via the epigenetic regulation of cholesterol trafficking. ADK inhibition is a promising approach for the treatment of atherosclerotic diseases.


ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , Adenosine Kinase/deficiency , Aorta/enzymology , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Epigenesis, Genetic , Foam Cells/enzymology , Mice, Knockout, ApoE , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Adenosine Kinase/genetics , Animals , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Cholesterol/metabolism , DNA Methylation , Disease Models, Animal , Female , Foam Cells/pathology , Male , Mice, Inbred C57BL , Plaque, Atherosclerotic , Signal Transduction
6.
Stem Cells Transl Med ; 7(6): 477-486, 2018 06.
Article En | MEDLINE | ID: mdl-29589874

As a powerful regulator of cellular homeostasis and metabolism, adenosine is involved in diverse neurological processes including pain, cognition, and memory. Altered adenosine homeostasis has also been associated with several diseases such as depression, schizophrenia, or epilepsy. Based on its protective properties, adenosine has been considered as a potential therapeutic agent for various brain disorders. Since systemic application of adenosine is hampered by serious side effects such as vasodilatation and cardiac suppression, recent studies aim at improving local delivery by depots, pumps, or cell-based applications. Here, we report on the characterization of adenosine-releasing human embryonic stem cell-derived neuroepithelial stem cells (long-term self-renewing neuroepithelial stem [lt-NES] cells) generated by zinc finger nuclease (ZFN)-mediated knockout of the adenosine kinase (ADK) gene. ADK-deficient lt-NES cells and their differentiated neuronal and astroglial progeny exhibit substantially elevated release of adenosine compared to control cells. Importantly, extensive adenosine release could be triggered by excitation of differentiated neuronal cultures, suggesting a potential activity-dependent regulation of adenosine supply. Thus, ZFN-modified neural stem cells might serve as a useful vehicle for the activity-dependent local therapeutic delivery of adenosine into the central nervous system. Stem Cells Translational Medicine 2018;7:477-486.


Adenosine/metabolism , Gene Editing/methods , Neural Stem Cells/metabolism , Neurons/metabolism , Adenosine/analysis , Adenosine Kinase/deficiency , Adenosine Kinase/genetics , Animals , Cell Line , Chromatography, High Pressure Liquid , Human Embryonic Stem Cells/cytology , Humans , Karyotyping , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/cytology , Neural Stem Cells/transplantation , Neurons/cytology , Polymorphism, Single Nucleotide , Zinc Finger Nucleases/genetics
7.
J Neurosci ; 36(48): 12117-12128, 2016 11 30.
Article En | MEDLINE | ID: mdl-27903722

Adenosine kinase (ADK) deficiency in human patients (OMIM:614300) disrupts the methionine cycle and triggers hypermethioninemia, hepatic encephalopathy, cognitive impairment, and seizures. To identify whether this neurological phenotype is intrinsically based on ADK deficiency in the brain or if it is secondary to liver dysfunction, we generated a mouse model with a brain-wide deletion of ADK by introducing a Nestin-Cre transgene into a line of conditional ADK deficient Adkfl/fl mice. These AdkΔbrain mice developed a progressive stress-induced seizure phenotype associated with spontaneous convulsive seizures and profound deficits in hippocampus-dependent learning and memory. Pharmacological, biochemical, and electrophysiological studies suggest enhanced adenosine levels around synapses resulting in an enhanced adenosine A1 receptor (A1R)-dependent protective tone despite lower expression levels of the receptor. Theta-burst-induced LTP was enhanced in the mutants and this was dependent on adenosine A2A receptor (A2AR) and tropomyosin-related kinase B signaling, suggesting increased activation of these receptors in synaptic plasticity phenomena. Accordingly, reducing adenosine A2A receptor activity in AdkΔbrain mice restored normal associative learning and contextual memory and attenuated seizure risk. We conclude that ADK deficiency in the brain triggers neuronal adaptation processes that lead to dysregulated synaptic plasticity, cognitive deficits, and increased seizure risk. Therefore, ADK mutations have an intrinsic effect on brain physiology and may present a genetic risk factor for the development of seizures and learning impairments. Furthermore, our data show that blocking A2AR activity therapeutically can attenuate neurological symptoms in ADK deficiency. SIGNIFICANCE STATEMENT: A novel human genetic condition (OMIM #614300) that is based on mutations in the adenosine kinase (Adk) gene has been discovered recently. Affected patients develop hepatic encephalopathy, seizures, and severe cognitive impairment. To model and understand the neurological phenotype of the human mutation, we generated a new conditional knock-out mouse with a brain-specific deletion of Adk (AdkΔbrain). Similar to ADK-deficient patients, AdkΔbrain mice develop seizures and cognitive deficits. We identified increased basal synaptic transmission and enhanced adenosine A2A receptor (A2AR)-dependent synaptic plasticity as the underlying mechanisms that govern these phenotypes. Our data show that neurological phenotypes in ADK-deficient patients are intrinsic to ADK deficiency in the brain and that blocking A2AR activity therapeutically can attenuate neurological symptoms in ADK deficiency.


Adenosine Kinase/deficiency , Adenosine/metabolism , Brain/physiopathology , Neuronal Plasticity , Receptor, Adenosine A2A/metabolism , Synaptic Transmission , Adenosine Kinase/genetics , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurotransmitter Agents/metabolism , Synapses/enzymology
8.
Neuroradiology ; 58(7): 697-703, 2016 Jul.
Article En | MEDLINE | ID: mdl-26993811

INTRODUCTION: Adenosine kinase deficiency (ADK deficiency) is a recently described disorder of methionine and adenosine metabolism resulting in a neurological phenotype with developmental delay, muscular hypotonia, and epilepsy as well as variable systemic manifestations. The underlying neuropathology is poorly understood. We have investigated MRI and (1)H-MRS changes in ADK deficiency in order to better understand the in vivo neuropathologic changes of ADK deficiency. METHODS: Systematic evaluation of 21 MRIs from eight patients (age range 9 days-14.6 years, mean 3.9 years, median 2.7 years) including diffusion-weighted imaging in six and (1)H-MRS in five patients. RESULTS: Brain maturation was delayed in the neonatal period and in infancy (6/6), but ultimately complete. White matter changes occurring in five of eight patients were discrete, periventricular, and unspecific (4/5), or diffuse with sparing of optic radiation, corona radiata, and pyramidal tracts (1/5). Choline was low in white matter spectra (3/3), while there was no indication of low creatine in white matter or basal ganglia (5/5), and diffusion was variably decreased or increased. Central tegmental tract hyperintensity was a common finding (6/8), as was supratentorial atrophy (6/8). CONCLUSIONS: MRI changes in ADK deficiency consist of delayed but ultimately completed brain maturation with later onset of mostly unspecific white matter changes and potentially transient central tegmental tract hyperintensity. Immaturity on neonatal MRI is consistent with prenatal onset of disease and reduced choline with lower membrane turnover resulting in delayed myelination and deficient myelin maintenance.


Adenosine Kinase/deficiency , Brain Diseases, Metabolic/enzymology , Brain Diseases, Metabolic/pathology , Brain/metabolism , Brain/pathology , Magnetic Resonance Imaging/methods , Proton Magnetic Resonance Spectroscopy/methods , Adenosine Kinase/metabolism , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Molecular Imaging/methods , Reproducibility of Results , Sensitivity and Specificity
9.
J Inherit Metab Dis ; 39(2): 273-83, 2016 Mar.
Article En | MEDLINE | ID: mdl-26642971

BACKGROUND: Adenosine kinase deficiency is a recently described defect affecting methionine metabolism with a severe clinical phenotype comprising mainly neurological and hepatic impairment and dysmorphism. METHODS: Clinical data of 11 additional patients from eight families with adenosine kinase deficiency were gathered through a retrospective questionnaire. Two liver biopsies of one patient were systematically evaluated. RESULTS: The main clinical symptoms are mild to severe liver dysfunction with neonatal onset, muscular hypotonia, global developmental retardation and dysmorphism (especially frontal bossing). Hepatic involvement is not a constant finding. Most patients have epilepsy and recurrent hypoglycemia due to hyperinsulinism. Major biochemical findings are intermittent hypermethioninemia, increased S-adenosylmethionine and S-adenosylhomocysteine in plasma and increased adenosine in urine. S-adenosylmethionine and S-adenosylhomocysteine are the most reliable biochemical markers. The major histological finding was pronounced microvesicular hepatic steatosis. Therapeutic trials with a methionine restricted diet indicate a potential beneficial effect on biochemical and clinical parameters in four patients and hyperinsulinism was responsive to diazoxide in two patients. CONCLUSION: Adenosine kinase deficiency is a severe inborn error at the cross-road of methionine and adenosine metabolism that mainly causes dysmorphism, brain and liver symptoms, but also recurrent hypoglycemia. The clinical phenotype varies from an exclusively neurological to a multi-organ manifestation. Methionine-restricted diet should be considered as a therapeutic option.


Adenosine Kinase/deficiency , Metabolic Diseases/mortality , Adenosine/metabolism , Adenosine/urine , Adenosine Kinase/metabolism , Adolescent , Adult , Biomarkers/blood , Biomarkers/metabolism , Biomarkers/urine , Child , Child, Preschool , Diet , Female , Humans , Hypoglycemia/metabolism , Hypoglycemia/mortality , Infant , Liver/metabolism , Liver/pathology , Liver Diseases/metabolism , Liver Diseases/mortality , Liver Diseases/pathology , Male , Metabolic Diseases/metabolism , Methionine/metabolism , Retrospective Studies , S-Adenosylhomocysteine/blood , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/blood , S-Adenosylmethionine/metabolism , Young Adult
10.
Appl Microbiol Biotechnol ; 97(3): 1183-90, 2013 Feb.
Article En | MEDLINE | ID: mdl-22790542

To isolate an S-adenosylmethionine (SAM)-accumulating yeast strain and to develop a more efficient method of producing SAM, we screened methionine-resistant strains using the yeast deletion library of budding yeast and isolated 123 strains. The SAM content in 81 of the 123 strains was higher than that in the parental strain BY4742. We identified ADO1 encoding adenosine kinase as one of the factors participating in high SAM accumulation. The X∆ado1 strain that was constructed from the X2180-1A strain (MAT a, ATCC 26786) could accumulate approximately 30-fold (18 mg/g dry cell weight) more SAM than the X2180-1A strain in yeast extract peptone dextrose medium. Furthermore, we attempted to identify the molecular basis underlying the differences in SAM accumulation between X∆ado1 and X2180-1A strains. DNA microarray analysis revealed that the genes involved in the methionine biosynthesis pathway, phosphate metabolism, and hexose transport were mainly overexpressed in the X∆ado1 strain compared with the X2180-1A strain. We also determined the levels of various metabolites involved in the methionine biosynthesis pathway and found increased content of SAM, tetrahydrofolate (THF), inorganic phosphate, polyphosphoric acid, and S-adenosylhomocysteine in the X∆ado1 strain. In contrast, the content of 5-methyl-THF, homocysteine, glutathione, and adenosine was decreased. These results indicated that the ∆ado1 strain could accumulate SAM because of preferential activation of the methionine biosynthesis pathway.


Adenosine Kinase/deficiency , Methionine/biosynthesis , S-Adenosylmethionine/metabolism , Saccharomyces cerevisiae/enzymology , Biosynthetic Pathways , Crosses, Genetic , Culture Media/chemistry , Gene Expression Profiling , Metabolome , Microarray Analysis , Saccharomyces cerevisiae/genetics
11.
Am J Hum Genet ; 89(4): 507-15, 2011 Oct 07.
Article En | MEDLINE | ID: mdl-21963049

Four inborn errors of metabolism (IEMs) are known to cause hypermethioninemia by directly interfering with the methionine cycle. Hypermethioninemia is occasionally discovered incidentally, but it is often disregarded as an unspecific finding, particularly if liver disease is involved. In many individuals the hypermethioninemia resolves without further deterioration, but it can also represent an early sign of a severe, progressive neurodevelopmental disorder. Further investigation of unclear hypermethioninemia is therefore important. We studied two siblings affected by severe developmental delay and liver dysfunction. Biochemical analysis revealed increased plasma levels of methionine, S-adenosylmethionine (AdoMet), and S-adenosylhomocysteine (AdoHcy) but normal or mildly elevated homocysteine (Hcy) levels, indicating a block in the methionine cycle. We excluded S-adenosylhomocysteine hydrolase (SAHH) deficiency, which causes a similar biochemical phenotype, by using genetic and biochemical techniques and hypothesized that there was a functional block in the SAHH enzyme as a result of a recessive mutation in a different gene. Using exome sequencing, we identified a homozygous c.902C>A (p.Ala301Glu) missense mutation in the adenosine kinase gene (ADK), the function of which fits perfectly with this hypothesis. Increased urinary adenosine excretion confirmed ADK deficiency in the siblings. Four additional individuals from two unrelated families with a similar presentation were identified and shown to have a homozygous c.653A>C (p.Asp218Ala) and c.38G>A (p.Gly13Glu) mutation, respectively, in the same gene. All three missense mutations were deleterious, as shown by activity measurements on recombinant enzymes. ADK deficiency is a previously undescribed, severe IEM shedding light on a functional link between the methionine cycle and adenosine metabolism.


Adenosine Kinase/deficiency , Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases/metabolism , Liver Diseases/pathology , Methionine/genetics , Methionine/metabolism , Adult , Amino Acid Metabolism, Inborn Errors/diagnosis , Brain Diseases/genetics , Child , Developmental Disabilities/genetics , Family Health , Female , Fibroblasts/metabolism , Homocysteine/blood , Homocysteine/genetics , Humans , Liver Diseases/genetics , Male , Methionine/blood , S-Adenosylhomocysteine/blood , S-Adenosylmethionine/blood , S-Adenosylmethionine/genetics
12.
J Neurosci ; 30(39): 13157-65, 2010 Sep 29.
Article En | MEDLINE | ID: mdl-20881134

Sleep and sleep intensity are enhanced by adenosine and its receptor agonists, whereas adenosine receptor antagonists induce wakefulness. Adenosine kinase (ADK) is the primary enzyme metabolizing adenosine in adult brain. To investigate whether adenosine metabolism or clearance affects sleep, we recorded sleep in mice with engineered mutations in Adk. Adk-tg mice overexpress a transgene encoding the cytoplasmic isoform of ADK in the brain but lack the nuclear isoform of the enzyme. Wild-type mice and Adk(+/-) mice that have a 50% reduction of the cytoplasmic and the nuclear isoforms of ADK served as controls. Adk-tg mice showed a remarkable reduction of EEG power in low frequencies in all vigilance states and in theta activity (6.25-11 Hz) in rapid eye movement (REM) sleep and waking. Adk-tg mice were awake 58 min more per day than wild-type mice and spent significantly less time in REM sleep (102 ± 3 vs 128 ± 3 min in wild type). After sleep deprivation, slow-wave activity (0.75-4 Hz), the intensity component of non-rapid eye movement sleep, increased significantly less in Adk-tg mice and their slow-wave energy was reduced. In contrast, the vigilance states and EEG spectra of Adk(+/-) and wild-type mice did not differ. Our data suggest that overexpression of the cytoplasmic isoform of ADK is sufficient to alter sleep physiology. ADK might orchestrate neurotransmitter pathways involved in the generation of EEG oscillations and regulation of sleep.


Adenosine Kinase/genetics , Sleep/genetics , Adenosine/antagonists & inhibitors , Adenosine/physiology , Adenosine Kinase/biosynthesis , Adenosine Kinase/deficiency , Animals , Cytoplasm/enzymology , Disease Models, Animal , Electroencephalography/methods , Isoenzymes/deficiency , Isoenzymes/genetics , Isoenzymes/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurotransmitter Agents/genetics , Neurotransmitter Agents/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Sleep/physiology , Sleep Deprivation/genetics , Sleep Deprivation/physiopathology
13.
Seizure ; 19(7): 390-6, 2010 Sep.
Article En | MEDLINE | ID: mdl-20688264

PURPOSE: Intracerebral delivery of anti-epileptic compounds represents a novel strategy for the treatment of refractory epilepsy. Adenosine is a possible candidate for local delivery based on its proven anti-epileptic effects. Neural stem cells constitute an ideal cell source for intracerebral transplantation and long-term drug delivery. In order to develop a cell-based system for the long-term delivery of adenosine, we isolated neural progenitor cells from adenosine kinase deficient mice (Adk(-/-)) and compared their differentiation potential and adenosine release properties with corresponding wild-type cells. METHODS: Fetal neural progenitor cells were isolated from the brains of Adk(-/-) and C57BL/6 mice fetuses and expanded in vitro. Before and after neural differentiation, supernatants were collected and assayed for adenosine release using liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS: Adk(-/-) cells secreted significantly more adenosine compared to wild-type cells at any time point of differentiation. Undifferentiated Adk(-/-) cells secreted 137+/-5 ng adenosine per 10(5) cells during 24 h in culture, compared to 11+/-1 ng released from corresponding wild-type cells. Adenosine release was maintained after differentiation as differentiated Adk(-/-) cells continued to release significantly more adenosine per 24 h (47+/-1 ng per 10(5) cells) compared to wild-type cells (3+/-0.2 ng per 10(5) cells). CONCLUSIONS: Fetal neural progenitor cells isolated from Adk(-/-) mice--but not those from C57BL/6 mice--release amounts of adenosine considered to be of therapeutic relevance.


Adenosine/administration & dosage , Astrocytes/transplantation , Epilepsy/drug therapy , Fetal Stem Cells/transplantation , Stem Cell Transplantation/methods , Adenosine Kinase/deficiency , Adenosine Kinase/genetics , Animals , Astrocytes/cytology , Astrocytes/metabolism , Blotting, Western , Cell Differentiation , Chromatography, Liquid , Female , Fetal Stem Cells/cytology , Fetal Stem Cells/metabolism , Immunohistochemistry , Injections, Intraventricular , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction
14.
Neuron Glia Biol ; 4(2): 91-9, 2008 May.
Article En | MEDLINE | ID: mdl-19674507

The astrocytic enzyme adenosine kinase (ADK) is a key negative regulator of the brain's endogenous anticonvulsant adenosine. Astrogliosis with concomitant upregulation of ADK is part of the epileptogenic cascade and contributes to seizure generation. To molecularly dissect the respective roles of astrogliosis and ADK-expression for seizure generation, we used a transgenic approach to uncouple ADK-expression from astrogliosis: in Adk-tg mice the endogenous Adk-gene was deleted and replaced by a ubiquitously expressed Adk-transgene with novel ectopic expression in pyramidal neurons, resulting in spontaneous seizures. Here, we followed a unique approach to selectively injure the CA3 of these Adk-tg mice. Using this strategy, we had the opportunity to study astrogliosis and epileptogenesis in the absence of the endogenous astrocytic Adk-gene. After triggering epileptogenesis we demonstrate astrogliosis without upregulation of ADK, but lack of seizures, whereas matching wild-type animals developed astrogliosis with upregulation of ADK and spontaneous recurrent seizures. By uncoupling ADK-expression from astrogliosis, we demonstrate that global expression levels of ADK rather than astrogliosis per se contribute to seizure generation.


Adenosine Kinase/metabolism , Astrocytes , Epilepsy/etiology , Gliosis/complications , Adenosine Kinase/deficiency , Adenosine Kinase/genetics , Animals , Astrocytes/enzymology , Brain/enzymology , Cell Death , Chronic Disease , Epilepsy/enzymology , Gliosis/enzymology , Kainic Acid , Male , Mice , Mice, Knockout , Mice, Transgenic , Pyramidal Cells/enzymology , Recurrence , Seizures/chemically induced , Seizures/etiology , Seizures/physiopathology , Seizures/prevention & control , Severity of Illness Index , Status Epilepticus/complications , Status Epilepticus/physiopathology , Time Factors , Tissue Distribution , Transgenes , Up-Regulation
15.
Biomaterials ; 27(26): 4599-607, 2006 Sep.
Article En | MEDLINE | ID: mdl-16709437

Adenosine kinase deficient (Adk-/-) embryonic stem cells (ESCs) encapsulated in synthetic polymers have previously been shown to provide therapeutic adenosine release and transient seizure suppression in epileptic rats. Here we explored the utility of biopolymer-substrates to promote long-term adenosine release from Adk-/- ESCs. Three different substrates were studied: (1) type I collagen (Col-1), (2) silk-fibroin (SF), and (3) poly(L-ornithine) (PO) coated tissue culture plastic. Adk-/- or wild type (wt) ESC-derived glial precursor cells were seeded on the substrates and cultured either in proliferation medium containing growth factors or in differentiation medium devoid of growth factors. In proliferation medium cell proliferation was higher and metabolic activity lower on Col-1 and PO substrates as compared to SF. Cells from both genotypes readily differentiated into astrocytes after growth factor removal on all substrates. Adk-/- cells cultured on biopolymers released significantly more adenosine than their wt counterparts at all developmental stages. Adenosine release was similar on SF and PO substrates and the amounts released from Adk-/- cells (>20 ng/ml) were considered to be of therapeutic relevance. Taken together, these results suggest that silk matrices are particularly suitable biomaterials for ESC encapsulation and for the design of adenosine releasing bioincubators for the treatment of epilepsy.


Adenosine Kinase/deficiency , Adenosine/metabolism , Biocompatible Materials/metabolism , Epilepsy/drug therapy , Fibroins/metabolism , Stem Cells/metabolism , Adenosine/therapeutic use , Adenosine Kinase/genetics , Animals , Capsules , Cell Differentiation , Cell Proliferation , Cells, Cultured , Collagen Type I/metabolism , Delayed-Action Preparations , Embryo, Mammalian/cytology , Glucose/metabolism , Hydrophobic and Hydrophilic Interactions , Mice , Mutation , Neuroglia/cytology , Neuroglia/enzymology , Neuroglia/metabolism , Peptides/metabolism , Stem Cells/cytology , Stem Cells/enzymology
16.
FEBS J ; 272(20): 5278-90, 2005 Oct.
Article En | MEDLINE | ID: mdl-16218958

This article is devoted to the study of redundancy and yield of salvage pathways in human erythrocytes. These cells are not able to synthesize ATP de novo. However, the salvage (recycling) of certain nucleosides or bases to give nucleotide triphosphates is operative. As the salvage pathways use enzymes consuming ATP as well as enzymes producing ATP, it is not easy to see whether a net synthesis of ATP is possible. As for pathways using adenosine, a straightforward assumption is that these pathways start with adenosine kinase. However, a pathway bypassing this enzyme and using S-adenosylhomocysteine hydrolase instead was reported. So far, this route has not been analysed in detail. Using the concept of elementary flux modes, we investigate theoretically which salvage pathways exist in erythrocytes, which enzymes belong to each of these and what relative fluxes these enzymes carry. Here, we compute the net overall stoichiometry of ATP build-up from the recycled substrates and show that the network has considerable redundancy. For example, four different pathways of adenine salvage and 12 different pathways of adenosine salvage are obtained. They give different ATP/glucose yields, the highest being 3:10 for adenine salvage and 2:3 for adenosine salvage provided that adenosine is not used as an energy source. Implications for enzyme deficiencies are discussed.


Adenine/metabolism , Adenosine Triphosphate/metabolism , Adenosine/metabolism , Adenosylhomocysteinase/metabolism , Erythrocytes/metabolism , Models, Biological , ADP Ribose Transferases/deficiency , ADP Ribose Transferases/metabolism , Adenosine Deaminase/deficiency , Adenosine Deaminase/metabolism , Adenosine Kinase/deficiency , Adenosine Kinase/metabolism , Humans , Metabolic Diseases/metabolism , Purine-Nucleoside Phosphorylase/deficiency , Purine-Nucleoside Phosphorylase/metabolism
17.
Brain ; 128(Pt 10): 2383-95, 2005 Oct.
Article En | MEDLINE | ID: mdl-15930047

Adenosine kinase (ADK) is considered to be the key regulator of the brain's endogenous anticonvulsant, adenosine. In adult brain, ADK is primarily expressed in a subpopulation of astrocytes and striking upregulation of ADK in these cells has been associated with astrogliosis after kainic acid-induced status epilepticus (KASE) in the kainic acid mouse model of temporal lobe epilepsy. To investigate the causal relationship between KASE-induced astrogliosis, upregulation of ADK and seizure activity, we have developed a novel mouse model [the Adktm1(-/-)-Tg(UbiAdk) mouse] lacking the endogenous astrocytic enzyme due to a targeted disruption of the endogenous gene, but containing an Adk transgene under the control of a human ubiquitin promoter. Mutant Adktm1(-/-)-Tg(UbiAdk) mice were characterized by increased brain ADK activity and constitutive overexpression of transgenic ADK throughout the brain, with particularly high levels in hippocampal pyramidal neurons. This ADK overexpression was associated with increased baseline levels of locomotion. Most importantly, two-thirds of the mutant mice analysed exhibited spontaneous seizure activity in the hippocampus and cortex. This was the direct consequence of transgene expression, since this seizure activity could be prevented by systemic application of the ADK inhibitor 5-iodotubercidin. Intrahippocampal injection of kainate in the mutant mice resulted in astrogliosis to the same extent as that observed in wild-type mice despite the absence of endogenous astrocytic ADK. Therefore, KASE-induced upregulation of endogenous ADK in wild-type mice is a consequence of astrogliosis. However, seizures in kainic acid-injected mutants displayed increased intra-ictal spike frequency compared with wild-type mice, indicating that, once epilepsy is established, increased levels of ADK aggravate seizure severity. We therefore conclude that therapeutic strategies that augment the adenosine system after astrogliosis-induced upregulation of ADK constitute a neurochemical rationale for the prevention of seizures in epilepsy.


Adenosine Kinase/genetics , Epilepsy, Temporal Lobe/genetics , Gliosis/genetics , Adenosine Kinase/deficiency , Animals , Astrocytes/physiology , Behavior, Animal , Brain/enzymology , Cerebral Cortex/physiopathology , Disease Models, Animal , Electroencephalography/methods , Enzyme Inhibitors/pharmacology , Epilepsy, Temporal Lobe/complications , Gliosis/complications , Gliosis/enzymology , Hippocampus/physiopathology , Kainic Acid , Locomotion , Male , Mice , Mice, Transgenic , Neurons/metabolism , Transgenes/genetics , Tubercidin/analogs & derivatives , Tubercidin/pharmacology , Up-Regulation
18.
Neurosci Lett ; 370(2-3): 160-5, 2004 Nov 11.
Article En | MEDLINE | ID: mdl-15488315

Based on the anticonvulsant and neuroprotective properties of adenosine, and based on the long-term survival potential of stem cell derived brain implants, adenosine releasing stem cells may constitute a novel tool for the treatment of epilepsy. Pluripotency and unlimited self-renewal make embryonic stem (ES) cells a particularly versatile donor source for cell transplantation. With the aim to test the feasibility of a stem cell-based delivery system for adenosine, both alleles of adenosine kinase (ADK), the major adenosine-metabolizing enzyme, were disrupted by homologous recombination in ES cells. Adk-/- ES cells were subjected to a glial differentiation protocol and, as a result, gave rise to proliferating glial precursors, which could be further differentiated into mature astrocytes and oligodendrocytes. Thus, a lack of ADK does not compromise the glial differentiation potential of ES cells. The Adk-/- ES cells yielded glial populations with an adenosine release of up to 40.1 +/- 6.0 ng per 10(5) cells per hour, an amount considered to be sufficient for seizure suppression. Our findings indicate that Adk-/- ES cells constitute a potential source for therapeutic adenosine releasing grafts.


Adenosine/metabolism , Neuroglia/metabolism , Stem Cell Transplantation , Stem Cells/physiology , Adenosine Kinase/deficiency , Adenosine Kinase/genetics , Analysis of Variance , Animals , Blotting, Western/methods , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Chromosome Mapping/methods , Embryo, Mammalian , Fluorescent Antibody Technique/methods , Gene Expression Regulation, Developmental/drug effects , Genetic Engineering , Glial Fibrillary Acidic Protein/immunology , Growth Substances/pharmacology , Intermediate Filament Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Nestin , Neuroglia/drug effects , O Antigens/metabolism , Oligodendroglia/metabolism , Polymerase Chain Reaction/methods
19.
J Med Chem ; 47(8): 1987-96, 2004 Apr 08.
Article En | MEDLINE | ID: mdl-15055998

Toxoplasma gondii is the most common cause of secondary CNS infections in immunocompromised persons such as AIDS patients. The major route of adenosine metabolism in T. gondii is direct phosphorylation to adenosine 5'-monophosphate (AMP) catalyzed by the enzyme adenosine kinase (EC 2.7.1.20). Adenosine kinase in T. gondii is significantly more active than any other purine salvage enzyme in this parasite and has been established as a potential chemotherapeutic target for the treatment of toxoplasmosis. Subversive substrates of T. gondii,but not the human, adenosine kinase are preferentially metabolized to their monophosphorylated forms and become selectively toxic to the parasites but not their host. 6-Benzylthioinosine (BTI) was identified as an excellent subversive substrate of T. gondii adenosine kinase. Herein, we report the synthesis of new analogues of BTI as subversive substrates for T. gondii adenosine kinase. These new subversive substrates were synthesized starting from tribenzoyl protected d-ribose. To accomplish the lead optimization process, a divergent and focused combinatorial library was synthesized using a polymer-supported trityl group at the 5'-position. The combinatorial library of 20 compounds gave several compounds more active than BTI. Structure-activity relationship studies showed that substitution at the para position plays a crucial role. To investigate the reasons for this discrimination, substrates with different substituents at the para position were studied by molecular modeling using Monte Carlo Conformational Search followed by energy minimization of the enzyme-ligand complex.


Adenosine Kinase/metabolism , Thioinosine/chemical synthesis , Toxoplasma/enzymology , Adenosine Kinase/chemistry , Adenosine Kinase/deficiency , Animals , Cells, Cultured , Coccidiostats/chemical synthesis , Coccidiostats/chemistry , Coccidiostats/pharmacology , Combinatorial Chemistry Techniques , Humans , Models, Molecular , Structure-Activity Relationship , Thioinosine/analogs & derivatives , Thioinosine/chemistry , Thioinosine/pharmacology , Toxoplasma/drug effects
20.
J Neurosci Methods ; 120(1): 85-94, 2002 Oct 15.
Article En | MEDLINE | ID: mdl-12351209

Adenosine is an efficient inhibitor of neuronal activity with the ability to suppress seizure activity in various animal models of epilepsy. In the present study adenosine-releasing neuronal cells were generated as a potential source for therapeutically active grafts. Mice with a genetic disruption of the gene encoding adenosine kinase (Adk(-/-))-the major adenosine metabolizing enzyme-were used as a source for the derivation of adenosine releasing neuronal cells. Since homozygous Adk(-/-) mice constitute a lethal phenotype, embryonic neuroectoderm was derived from intercrosses of Adk(+/-)-mice. Therefore, a rapid genotyping procedure had to be developed using a fluorescent 5'-exonuclease (TaqMan) assay, which permitted the genotyping of embryonic cell material within 3 h. During this time period the cells to be grafted displayed an unaltered viability. Cultured neuroectodermal Adk(-/-) cells released up to 2 micro g adenosine per mg protein per hour. Adk(-/-) neuroectoderm grafted into the lateral brain ventricle of adult mice was found to survive for at least 6 weeks. The method described here suggests the feasibility to graft adenosine releasing neuroectodermal cells as a potential therapeutic approach for the treatment of pharmacoresistant epilepsy.


Adenosine Kinase/deficiency , Brain Tissue Transplantation/methods , Ectoderm/transplantation , Fluorescent Dyes , Neurons/transplantation , Polymerase Chain Reaction/methods , Adenosine Kinase/genetics , Animals , Cells, Cultured , Embryo, Mammalian , Genotype , Lateral Ventricles/transplantation , Mice , Mice, Mutant Strains , Mice, Transgenic , Transplants
...