Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Sci Rep ; 12(1): 13825, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35970890

ABSTRACT

Infantile neuroaxonal dystrophy (INAD) is a rare paediatric neurodegenerative condition caused by mutations in the PLA2G6 gene, which is also the causative gene for PARK14-linked young adult-onset dystonia parkinsonism. INAD patients usually die within their first decade of life, and there are currently no effective treatments available. GLP1 receptor (GLP-1R) agonists are licensed for treating type 2 diabetes mellitus but have also demonstrated neuroprotective properties in a clinical trial for Parkinson's disease. Therefore, we evaluated the therapeutic efficacy of a new recently licensed GLP-1R agonist diabetes drug in a mouse model of INAD. Systemically administered high-dose semaglutide delivered weekly to juvenile INAD mice improved locomotor function and extended the lifespan. An investigation into the mechanisms underlying these therapeutic effects revealed that semaglutide significantly increased levels of key neuroprotective molecules while decreasing those involved in pro-neurodegenerative pathways. The expression of mediators in both the apoptotic and necroptotic pathways were also significantly reduced in semaglutide treated mice. A reduction of neuronal loss and neuroinflammation was observed. Finally, there was no obvious inflammatory response in wild-type mice associated with the repeated high doses of semaglutide used in this study.


Subject(s)
Diabetes Mellitus, Type 2 , Neuroaxonal Dystrophies , Parkinsonian Disorders , Animals , Disease Models, Animal , Dystonic Disorders , Group VI Phospholipases A2/deficiency , Mice , Neuroaxonal Dystrophies/genetics , Parkinsonian Disorders/genetics
2.
Parkinsonism Relat Disord ; 101: 66-74, 2022 08.
Article in English | MEDLINE | ID: mdl-35803092

ABSTRACT

BACKGROUND: PLA2G6-Associated Neurodegeneration, PLAN, is subdivided into: Infantile neuroaxonal dystrophy, atypical neuroaxonal dystrophy, and adult-onset dystonia parkinsonism [1]. It is elicited by a biallelic pathogenic variant in phospholipase A2 group VI (PLA2G6) gene. In this study we describe new cases and provide a comprehensive review of previously published cases. METHODS: Eleven patients, from four different institutions and four different countries. All underwent a comprehensive chart review. RESULTS: Ages at onset ranged from 1 to 36 years, with a median of 16 and a mean of 16.18 ± 11.91 years. Phenotypic characteristics were heterogenous and resembled that of patients with infantile neuroaxonal dystrophy (n = 2), atypical neuroaxonal dystrophy (n = 1), adult-onset dystonia parkinsonism (n = 1), complex hereditary spastic paraparesis (n = 3), and early onset Parkinson's disease (n = 2). Parental genetic studies were performed for all patients and confirmed with sanger sequencing in five. Visual evoked potential illustrated optic atrophy in P4. Mineralization was evident in brain magnetic resonance imaging of P1, P2, P4, P5, P7, and P11. Single photon emission computed tomography was conducted for three patients, revealed decreased perfusion in the occipital lobes for P10. DaTscan was performed for P11 and showed decreased uptake in the deep gray matter, bilateral caudate nuclei, and bilateral putamen. Positive response to Apomorphine was noted for P10 and to Baclofen in P2, and P3. CONCLUSIONS: PLAN encompasses a wide clinical spectrum. Age and symptom at onset are crucial when classifying patients. Reporting new variants is critical to draw more attention to this condition and identify biomarkers to arrive at potential therapeutics.


Subject(s)
Dystonic Disorders , Neuroaxonal Dystrophies , Parkinsonian Disorders , Adolescent , Adult , Child , Child, Preschool , Evoked Potentials, Visual , Group VI Phospholipases A2/deficiency , Group VI Phospholipases A2/genetics , Humans , Infant , Iron Metabolism Disorders , Mutation , Neuroaxonal Dystrophies/diagnostic imaging , Neuroaxonal Dystrophies/genetics , Parkinsonian Disorders/diagnostic imaging , Parkinsonian Disorders/genetics , Phenotype , Young Adult
3.
Mol Genet Genomics ; 296(1): 235-242, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33159255

ABSTRACT

Sporadic occurrences of neurodegenerative disorders including neuroaxonal dystrophy (NAD) have been previously reported in sheep. However, so far no causative genetic variant has been found for ovine NAD. The aim of this study was to characterize the phenotype and the genetic aetiology of an early-onset neurodegenerative disorder observed in several lambs of purebred Swaledale sheep, a native English breed. Affected lambs showed progressive ataxia and stiff gait and subsequent histopathological analysis revealed the widespread presence of axonal spheroid indicating neuronal degeneration. Thus, the observed clinical phenotype could be explained by a novel form of NAD. After SNP genotyping and subsequent linkage mapping within a paternal half-sib pedigree with a total of five NAD-affected lambs, we identified two loss-of-function variants by whole-genome sequencing in the ovine PLA2G6 gene situated in a NAD-linked genome region on chromosome 3. All cases were carriers of a compound heterozygous splice site variant in intron 2 and a nonsense variant in exon 8. Herein we present evidence for the occurrence of a familial novel form of recessively inherited NAD in sheep due to allelic heterogeneity at PLA2G6. This study reports two pathogenic variants in PLA2G6 causing a novel form of NAD in Swaledale sheep which enables selection against this fatal disorder.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Group VI Phospholipases A2/genetics , Neuroaxonal Dystrophies/genetics , Neuroaxonal Dystrophies/veterinary , Polymorphism, Single Nucleotide , Sheep Diseases/genetics , Alternative Splicing , Amyloid beta-Protein Precursor/metabolism , Animals , Base Sequence , Chromosome Mapping , Chromosomes, Mammalian/chemistry , Exons , Female , Gene Expression , Genetic Linkage , Group VI Phospholipases A2/deficiency , Heterozygote , Introns , Male , Neuroaxonal Dystrophies/metabolism , Neuroaxonal Dystrophies/pathology , Sheep , Sheep Diseases/metabolism , Sheep Diseases/pathology , Sheep, Domestic , Whole Genome Sequencing
5.
Mol Neurobiol ; 55(8): 6734-6754, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29344929

ABSTRACT

This study aimed to gain insights into the pathophysiology underlying PLA2G6-associated neurodegeneration that is implicated in three different neurological disorders, suggesting that other, unknown genetic or environmental factors might contribute to its wide phenotypic expression. To accomplish this, we downregulated the function of pla2g6 in the zebrafish nervous system, performed parkinsonism-related phenotypic characterization, and determined the effects of gene regulation upon the loss of pla2g6 function by using RNA sequencing and downstream analyses. Pla2g6 deficiency resulted in axonal degeneration, dopaminergic and motor neuron cell loss, and increased ß-synuclein expression. We also observed that many of the identified, differentially expressed genes were implicated in other brain disorders, which might explain the variable phenotypic expression of pla2g6-associated disease, and found that top enriched canonical pathways included those already known or suggested to play a major role in the pathogenesis of Parkinson's disease. Our data support that pla2g6 is relevant for cranial motor development with significant implications in the pathophysiology underlying Parkinson's disease.


Subject(s)
Apoptosis , Axons/pathology , Brain/pathology , Dopaminergic Neurons/pathology , Group VI Phospholipases A2/deficiency , Nerve Degeneration/pathology , Zebrafish Proteins/deficiency , Zebrafish/metabolism , beta-Synuclein/metabolism , Animals , Apoptosis/drug effects , Axons/drug effects , Axons/metabolism , Base Sequence , Body Patterning/drug effects , Brain/drug effects , Brain/metabolism , Conserved Sequence , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/drug effects , Group VI Phospholipases A2/genetics , Group VI Phospholipases A2/metabolism , Humans , Larva/drug effects , Larva/genetics , Larva/growth & development , Morpholinos/pharmacology , Motor Neurons/drug effects , Motor Neurons/metabolism , Motor Neurons/pathology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Zebrafish/embryology , Zebrafish Proteins/genetics
6.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(12): 1520-1533, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28888832

ABSTRACT

Ageing is a major risk factor for various forms of liver and gastrointestinal (GI) disease and genetic background may contribute to the pathogenesis of these diseases. Group VIA phospholipase A2 or iPLA2ß is a homeostatic PLA2 by playing a role in phospholipid metabolism and remodeling. Global iPLA2ß-/- mice exhibit aged-dependent phenotypes with body weight loss and abnormalities in the bone and brain. We have previously reported the abnormalities in these mutant mice showing susceptibility for chemical-induced liver injury and colitis. We hypothesize that iPLA2ß deficiency may sensitize with ageing for an induction of GI injury. Male wild-type and iPLA2ß-/- mice at 4 and 20-22months of age were studied. Aged, but not young, iPLA2ß-/-mice showed increased hepatic fibrosis and biliary ductular expansion as well as severe intestinal atrophy associated with increased apoptosis, pro-inflammation, disrupted tight junction, and reduced number of mucin-containing globlet cells. This damage was associated with decreased expression of intestinal endoplasmic stress XBP1 and its regulator HNF1α, FATP4, ACSL5, bile-acid transport genes as well as nuclear receptors LXRα and FXR. By LC/MS-MS profiling, iPLA2ß deficiency in aged mice caused an increase of intestinal arachidonate-containing phospholipids concomitant with a decrease in ceramides. By the suppression of intestinal FXR/FGF-15 signaling, hepatic bile-acid synthesis gene expression was increased leading to an elevation of secondary and hydrophobic bile acids in liver, bile, and intestine. In conclusions, ageing sensitized by iPLA2ß deficiency caused a decline of key intestinal homeostatic genes resulting in the development of GI disease in a gut-to-liver manner.


Subject(s)
Aging/metabolism , Bile Acids and Salts/metabolism , Ceramides/metabolism , Group VI Phospholipases A2/deficiency , Intestinal Diseases/metabolism , Liver Cirrhosis/metabolism , Phospholipids/metabolism , Aging/genetics , Aging/pathology , Animals , Bile Acids and Salts/genetics , Ceramides/genetics , Intestinal Diseases/genetics , Intestinal Diseases/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Mice , Mice, Knockout , Phospholipids/genetics
7.
Biochim Biophys Acta ; 1861(5): 449-61, 2016 May.
Article in English | MEDLINE | ID: mdl-26873633

ABSTRACT

PLA2G6 or GVIA calcium-independent PLA2 (iPLA2ß) is identified as one of the NAFLD modifier genes in humans, and thought to be a target for NAFLD therapy. iPLA2ß is known to play a house-keeping role in phospholipid metabolism and remodeling. However, its role in NAFLD pathogenesis has not been supported by results obtained from high-fat feeding of iPLA2ß-null (PKO) mice. Unlike livers of human NAFLD and genetically obese rodents, fatty liver induced by high-fat diet is not associated with depletion of hepatic phospholipids. We therefore tested whether iPLA2ß could regulate obesity and hepatic steatosis in leptin-deficient mice by cross-breeding PKO with ob/ob mice to generate ob/ob-PKO mice. Here we observed an improvement in ob/ob-PKO mice with significant reduction in serum enzymes, lipids, glucose, insulin as well as improved glucose tolerance, and reduction in islet hyperplasia. The improvement in hepatic steatosis measured by liver triglycerides, fatty acids and cholesterol esters was associated with decreased expression of PPARγ and de novo lipogenesis genes, and the reversal of ß-oxidation gene expression. Notably, ob/ob livers contained depleted levels of lysophospholipids and phospholipids, and iPLA2ß deficiency in ob/ob-PKO livers lowers the former, but replenished the latter particularly phosphatidylethanolamine (PE) and phosphatidylcholine (PC) that contained arachidonic (AA) and docosahexaenoic (DHA) acids. Compared with WT livers, PKO livers also contained increased PE and PC containing AA and DHA. Thus, iPLA2ß deficiency protected against obesity and ob/ob fatty liver which was associated with hepatic fatty-acyl phospholipid remodeling. Our results support the deleterious role of iPLA2ß in severe obesity associated NAFLD.


Subject(s)
Fatty Acids/blood , Group VI Phospholipases A2/deficiency , Liver/enzymology , Non-alcoholic Fatty Liver Disease/prevention & control , Obesity/prevention & control , Phospholipids/blood , Animals , Apoptosis , Arachidonic Acid/blood , Blood Glucose/metabolism , Cholesterol Esters/blood , Disease Models, Animal , Docosahexaenoic Acids/blood , Gene Expression Regulation , Genotype , Group VI Phospholipases A2/genetics , Insulin/blood , Insulin Resistance , Liver/pathology , Lysophospholipids/blood , Male , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Obesity/blood , Obesity/enzymology , Obesity/genetics , Obesity/pathology , Oxidation-Reduction , PPAR gamma/genetics , PPAR gamma/metabolism , Phenotype , Phosphatidylcholines/blood , Phosphatidylethanolamines/blood , Triglycerides/blood
8.
J Med Genet ; 53(3): 180-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26668131

ABSTRACT

BACKGROUND: Mutations in PLA2G6, which encodes the calcium-independent phospholipase A2 group VI, cause neurodegeneration and diffuse cortical Lewy body formation by a yet undefined mechanism. We assessed whether altered protein glycosylation due to abnormal Golgi morphology might be a factor in the pathology of this disease. METHODS: Three patients presented with PLA2G6-associated neurodegeneration (PLAN); two had infantile neuroaxonal dystrophy (INAD) and one had adult-onset dystonia-parkinsonism. We analysed protein N-linked and O-linked glycosylation in cerebrospinal fluid, plasma, urine, and cultured skin fibroblasts using high performance liquid chromatography (HPLC) and matrix-assisted laser desorption ionization--time of flight/mass spectrometry (MALDI-TOF/MS). We also assessed sialylation and Golgi morphology in cultured fibroblasts by immunofluorescence and performed rescue experiments using a lentiviral vector. RESULTS: The patients with INAD had PLA2G6 mutations NM_003560.2: c.[950G>T];[426-1077dup] and c.[1799G>A];[2221C>T] and the patient with dystonia-parkinsonism had PLA2G6 mutations NM_003560.2: c.[609G>A];[2222G>A]. All three patients had altered Golgi morphology and abnormalities of protein O-linked glycosylation and sialylation in cultured fibroblasts that were rescued by lentiviral overexpression of wild type PLA2G6. CONCLUSIONS: Our findings add altered Golgi morphology, O-linked glycosylation and sialylation defects to the phenotypical spectrum of PLAN; these pathways are essential for correct processing and distribution of proteins. Lewy body and Tau pathology, two neuropathological features of PLAN, could emerge from these defects. Therefore, Golgi morphology, O-linked glycosylation and sialylation may play a role in the pathogenesis of PLAN and perhaps other neurodegenerative disorders.


Subject(s)
Dystonic Disorders/metabolism , Dystonic Disorders/pathology , Golgi Apparatus/ultrastructure , Group VI Phospholipases A2/deficiency , Neuroaxonal Dystrophies/metabolism , Neuroaxonal Dystrophies/pathology , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Adult , Cells, Cultured , Dystonic Disorders/genetics , Female , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Glycosylation , Golgi Apparatus/metabolism , Group VI Phospholipases A2/genetics , Group VI Phospholipases A2/metabolism , Humans , Infant , Male , Mutation , Neuroaxonal Dystrophies/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Parkinsonian Disorders/genetics , Sialyltransferases/metabolism
9.
PLoS One ; 10(10): e0141629, 2015.
Article in English | MEDLINE | ID: mdl-26506412

ABSTRACT

Mutations in PLA2G6 have been proposed to be the cause of neurodegeneration with brain iron accumulation type 2. The present study aimed to clarify the mechanism underlying brain iron accumulation during the deficiency of calcium-independent phospholipase A2 beta (iPLA2ß), which is encoded by the PLA2G6 gene. Perl's staining with diaminobenzidine enhancement was used to visualize brain iron accumulation. Western blotting was used to investigate the expression of molecules involved in iron homeostasis, including divalent metal transporter 1 (DMT1) and iron regulatory proteins (IRP1 and 2), in the brains of iPLA2ß-knockout (KO) mice as well as in PLA2G6-knockdown (KD) SH-SY5Y human neuroblastoma cells. Furthermore, mitochondrial functions such as ATP production were examined. We have discovered for the first time that marked iron deposition was observed in the brains of iPLA2ß-KO mice since the early clinical stages. DMT1 and IRP2 were markedly upregulated in all examined brain regions of aged iPLA2ß-KO mice compared to age-matched wild-type control mice. Moreover, peroxidized lipids were increased in the brains of iPLA2ß-KO mice. DMT1 and IRPs were significantly upregulated in PLA2G6-KD cells compared with cells treated with negative control siRNA. Degeneration of the mitochondrial inner membrane and decrease of ATP production were observed in PLA2G6-KD cells. These results suggest that the genetic ablation of iPLA2ß increased iron uptake in the brain through the activation of IRP2 and upregulation of DMT1, which may be associated with mitochondrial dysfunction.


Subject(s)
Cation Transport Proteins/genetics , Group VI Phospholipases A2/genetics , Iron Regulatory Protein 2/genetics , Iron/metabolism , Animals , Brain/metabolism , Brain/pathology , Cation Transport Proteins/metabolism , Disease Models, Animal , Female , Group VI Phospholipases A2/deficiency , Group VI Phospholipases A2/metabolism , Humans , Iron Regulatory Protein 2/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/pathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Transcriptional Activation
10.
FEBS Lett ; 589(18): 2367-71, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26206229

ABSTRACT

Previous reports from our lab identified a mitochondrial calcium-independent phospholipase A2 activity that is activated when the mitochondrial membrane potential is decreased. This activity was demonstrated to influence occurrence of the permeability transition. Originally, this activity was ascribed to the iPLA2ß protein. Recently, both iPLA2ß and iPLA2γ knock out mice have been generated. It has been shown by others that the iPLA2γ plays a significant role in progression of the permeability transition. In this paper, using the iPLA2ß and iPLA2γ knock out mice we show that the membrane potential sensitive activity is the iPLA2γ.


Subject(s)
Group VI Phospholipases A2/metabolism , Membrane Potential, Mitochondrial , Mitochondria, Liver/metabolism , Animals , Enzyme Activation/drug effects , Gene Knockout Techniques , Group VI Phospholipases A2/antagonists & inhibitors , Group VI Phospholipases A2/deficiency , Group VI Phospholipases A2/genetics , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Naphthalenes/pharmacology , Pyrones/pharmacology , Rats
11.
Pediatr Neurol ; 51(6): 850-3, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25439493

ABSTRACT

BACKGROUND: Alternating hemiplegia of childhood and rapid-onset dystonia parkinsonism are two separate movement disorders with different dominant mutations in the same sodium-potassium transporter ATPase subunit gene, ATP1A3. PATIENT: We present a child with topiramate-responsive alternating hemiplegia of childhood who was tested for an ATP1A3 gene mutation. RESULTS: Gene sequencing revealed an identical ATP1A3 mutation as in three typical adult-onset rapid-onset dystonia parkinsonism cases but never previously described in an alternating hemiplegia of childhood case. CONCLUSION: The discordance of these phenotypes suggests that there are other undiscovered environmental, genetic, or epigenetic factors influencing the development of alternating hemiplegia of childhood or rapid-onset dystonia parkinsonism.


Subject(s)
Dystonic Disorders/genetics , Group VI Phospholipases A2/deficiency , Hemiplegia/genetics , Parkinsonian Disorders/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Adult , Child, Preschool , Dystonic Disorders/etiology , Female , Group VI Phospholipases A2/genetics , Hemiplegia/etiology , Humans , Male , Mutation , Parkinsonian Disorders/etiology
12.
PLoS One ; 9(10): e109409, 2014.
Article in English | MEDLINE | ID: mdl-25313821

ABSTRACT

In platelets, group IVA cytosolic phospholipase A2 (cPLA2α) has been implicated as a key regulator in the hydrolysis of platelet membrane phospholipids, leading to pro-thrombotic thromboxane A2 and anti-thrombotic 12-(S)-hydroxyeicosatetranoic acid production. However, studies using cPLA2α-deficient mice have indicated that other PLA2(s) may also be involved in the hydrolysis of platelet glycerophospholipids. In this study, we found that group VIB Ca2+-independent PLA2 (iPLA2γ)-deficient platelets showed decreases in adenosine diphosphate (ADP)-dependent aggregation and ADP- or collagen-dependent thromboxane A2 production. Electrospray ionization mass spectrometry analysis of platelet phospholipids revealed that fatty acyl compositions of ethanolamine plasmalogen and phosphatidylglycerol were altered in platelets from iPLA2γ-null mice. Furthermore, mice lacking iPLA2γ displayed prolonged bleeding times and were protected against pulmonary thromboembolism. These results suggest that iPLA2γ is an additional, long-sought-after PLA2 that hydrolyzes platelet membranes and facilitates platelet aggregation in response to ADP.


Subject(s)
Blood Platelets/metabolism , Group VI Phospholipases A2/metabolism , Adenosine Diphosphate/metabolism , Animals , Calcium/metabolism , Collagen/metabolism , Disease Susceptibility , Group VI Phospholipases A2/deficiency , Group VI Phospholipases A2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phospholipids/analysis , Platelet Activation , Platelet Aggregation , Receptors, Purinergic P2Y/metabolism , Serotonin/metabolism , Signal Transduction , Spectrometry, Mass, Electrospray Ionization , Thrombosis/metabolism , Thrombosis/pathology , Thromboxane A2/metabolism
13.
Neurochem Res ; 39(8): 1522-32, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24919816

ABSTRACT

Calcium-independent phospholipase A2 group VIa (iPLA2ß) preferentially releases docosahexaenoic acid (DHA) from the sn-2 position of phospholipids. Mutations of its gene, PLA2G6, are found in patients with several progressive motor disorders, including Parkinson disease. At 4 months, PLA2G6 knockout mice (iPLA2ß(-/-)) show minimal neuropathology but altered brain DHA metabolism. By 1 year, they develop motor disturbances, cerebellar neuronal loss, and striatal α-synuclein accumulation. We hypothesized that older iPLA2ß(-/-) mice also would exhibit inflammatory and other neuropathological changes. Real-time polymerase chain reaction and Western blotting were performed on whole brain homogenate from 15 to 20-month old male iPLA2ß(-/-) or wild-type (WT) mice. These older iPLA2ß(-/-) mice compared with WT showed molecular evidence of microglial (CD-11b, iNOS) and astrocytic (glial fibrillary acidic protein) activation, disturbed expression of enzymes involved in arachidonic acid metabolism, loss of neuroprotective brain derived neurotrophic factor, and accumulation of cytokine TNF-α messenger ribonucleic acid, consistent with neuroinflammatory pathology. There was no evidence of synaptic loss, of reduced expression of dopamine active reuptake transporter, or of accumulation of the Parkinson disease markers Parkin or Pink1. iPLA2γ expression was unchanged. iPLA2ß deficient mice show evidence of neuroinflammation and associated neuropathology with motor dysfunction in later life. These pathological biomarkers could be used to assess efficacy of dietary intervention, antioxidants or other therapies on disease progression in this mouse model of progressive human motor diseases associated with a PLA2G6 mutation.


Subject(s)
Aging/metabolism , Disease Models, Animal , Disease Progression , Group VI Phospholipases A2/deficiency , Motor Skills Disorders/metabolism , Aging/genetics , Animals , Group VI Phospholipases A2/genetics , Male , Mice , Mice, Knockout , Motor Skills Disorders/genetics , Motor Skills Disorders/pathology
14.
Histol Histopathol ; 28(8): 965-9, 2013 08.
Article in English | MEDLINE | ID: mdl-23467909

ABSTRACT

Infantile neuroaxonal dystrophy (INAD) is a severe neurodegenerative disease characterized by its early onset. PLA2G6, which encodes a phospholipase A2, iPLA2ß, has been identified as a causative gene of INAD. iPLA2ß has been shown to be involved in various physiological and pathological processes, including immunity, cell death, and cell membrane homeostasis. Gene targeted mice with a null mutation of Pla2g6 develop the INAD phenotype as late as approximately 1 to 2 years after birth. Recently, another INAD mouse model, Pla2g6-INAD mice line, has been established. The Pla2g6-INAD mice bear a point mutation in the ankyrin repeat domain of Pla2g6 generated by N-ethyl-N-nitrosourea mutagenesis. These mutant mice develop severe motor dysfunction and hematopoietic abnormality in a manner following Mendelian law. The mice showed the abnormal gait and poor performance as early as 7 to 8 weeks of age, detected by hanging grip test. Neuropathological examination revealed widespread formation of spheroids containing tubulovesicular membranes similar to human INAD. Molecular and biochemical analysis revealed that the mutant mice expressed Pla2g6 mRNA and protein, but the mutated Pla2g6 protein had no glycerophospholipid-catalyzing enzyme activity. When analyzed the offspring which bear Pla2g6 knockout allele and Pla2g6-INAD allele, abnormal gait appeared slightly later than Pla2g6-INAD homozygotes but with earlier onset than the Pla2g6 knockout homozygotes. This result suggests that mutant Pla2g6 protein contributes to early onset of INAD symptoms in the absence of intact Pla2g6 protein. The analysis of various INAD mouse models may help to understand the pathogenesis of neurodegenerative diseases, including INAD.


Subject(s)
Group VI Phospholipases A2/genetics , Neuroaxonal Dystrophies/genetics , Alleles , Animals , Disease Models, Animal , Group VI Phospholipases A2/deficiency , Hematopoiesis/genetics , Humans , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Mutation , Neuroaxonal Dystrophies/metabolism , Neurodegenerative Diseases/metabolism , Phospholipases A2/metabolism , Point Mutation
15.
Neurosci Lett ; 523(1): 35-8, 2012 Aug 08.
Article in English | MEDLINE | ID: mdl-22743658

ABSTRACT

Several causative genes have been identified for both dystonia-parkinsonism and neurodegeneration with brain iron accumulation (NBIA), yet many patients do not have mutations in any of the known genes. Mutations in the ATP13A2 lead to Kufor Rakeb disease, a form of autosomal recessive juvenile parkinsonism that also features oromandibular dystonia. More recently, evidence of iron deposition in the caudate and putamen have been reported in patients with ATP13A2 mutations. We set out to determine the frequency of ATP13A2 mutations in cohorts of idiopathic NBIA and dystonia-parkinsonism. We screened for large deletions using whole genome arrays, and sequenced the entire coding region in 92 cases of NBIA and 76 cases of dystonia-parkinsonism. A number of coding and non-coding sequence variants were identified in a heterozygous state, but none were predicted to be pathogenic based on in silico analyses. Our results indicate that ATP13A2 mutations are a rare cause of both NBIA and dystonia-parkinsonism.


Subject(s)
Dystonic Disorders/epidemiology , Dystonic Disorders/genetics , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Iron Metabolism Disorders/epidemiology , Iron Metabolism Disorders/genetics , Neuroaxonal Dystrophies/epidemiology , Neuroaxonal Dystrophies/genetics , Parkinsonian Disorders/epidemiology , Parkinsonian Disorders/genetics , Proton-Translocating ATPases/genetics , Adult , Aged , Aged, 80 and over , Cohort Studies , Comorbidity , Female , Genetic Markers/genetics , Group VI Phospholipases A2/deficiency , Group VI Phospholipases A2/genetics , Humans , Internationality , Male , Middle Aged , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Prevalence , Risk Factors , Young Adult
16.
Am J Physiol Lung Cell Mol Physiol ; 302(1): L47-55, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21984569

ABSTRACT

An early event in the pathogenesis of emphysema is the development of inflammation associated with accumulation of polymorphonuclear leukocytes (PMN) in small airways, and inflammatory cell recruitment from the circulation involves migration across endothelial and epithelial cell barriers. Platelet-activating factor (PAF) promotes transendothelial migration in several vascular beds, and we postulated that increased PAF production in the airways of smokers might enhance inflammatory cell recruitment and exacerbate inflammation. To examine this possibility, we incubated human lung microvascular endothelial cells (HMVEC-L) with cigarette smoke extract (CSE) and found that CSE inhibits PAF-acetylhydrolase (PAF-AH) activity. This enhances HMVEC-L PAF production and PMN adherence, and adherence is blocked by PAF receptor antagonists (CV3988 or ginkgolide B). CSE also inhibited PAF-AH activity of lung endothelial cells isolated from wild-type (WT) and iPLA(2)ß knockout mice, and with WT cells, CSE enhanced PAF production and RAW 264.7 cell adherence. In contrast, CSE did not affect PAF production or RAW 264.7 cell adherence to iPLA(2)ß-null cells, suggesting that iPLA(2)ß plays an important role in PAF production by lung endothelial cells. These findings suggest that inhibition of PAF-AH by components of cigarette smoke may initiate or exacerbate inflammatory lung disease by enhancing PAF production and promoting accumulation of inflammatory cells in small airways. In addition, iPLA(2)ß is identified as a potential target for therapeutic interventions to reduce airway inflammation and the progression of chronic lung disease.


Subject(s)
Endothelial Cells/metabolism , Group VI Phospholipases A2 , Platelet Activating Factor/metabolism , Tobacco Smoke Pollution/adverse effects , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Animals , Cell Adhesion/drug effects , Cell Line , Group VI Phospholipases A2/deficiency , Group VI Phospholipases A2/drug effects , Humans , Inflammation/drug therapy , Inflammation/physiopathology , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Neutrophils/metabolism , Phospholipid Ethers/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Pulmonary Disease, Chronic Obstructive/physiopathology , Receptors, G-Protein-Coupled/antagonists & inhibitors
17.
PLoS One ; 6(10): e26991, 2011.
Article in English | MEDLINE | ID: mdl-22046428

ABSTRACT

Infantile neuroaxonal dystrophy (INAD) is a progressive, autosomal recessive neurodegenerative disease characterized by axonal dystrophy, abnormal iron deposition and cerebellar atrophy. This disease was recently mapped to PLA2G6, which encodes group VI Ca(2+)-independent phospholipase A(2) (iPLA(2) or iPLA(2)ß). Here we show that genetic ablation of PLA2G6 in mice (iPLA(2)ß(-/-)) leads to the development of cerebellar atrophy by the age of 13 months. Atrophied cerebella exhibited significant loss of Purkinje cells, as well as reactive astrogliosis, the activation of microglial cells, and the pronounced up-regulation of the pro-inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß). Moreover, glial cell activation and the elevation in TNF-α and IL-1ß expression occurred before apparent cerebellar atrophy. Our findings indicate that the absence of PLA2G6 causes neuroinflammation and Purkinje cell loss and ultimately leads to cerebellar atrophy. Our study suggests that iPLA(2)ß(-/-) mice are a valuable model for cerebellar atrophy in INAD and that early anti-inflammatory therapy may help slow the progression of cerebellar atrophy in this deadly neurodegenerative disease.


Subject(s)
Atrophy/genetics , Cerebellar Diseases/pathology , Group VI Phospholipases A2/deficiency , Neuroglia/pathology , Purkinje Cells/pathology , Animals , Cerebellar Diseases/genetics , Group VI Phospholipases A2/genetics , Interleukin-1beta , Mice , Microglia , Neuroaxonal Dystrophies , Neuroglia/immunology , Tumor Necrosis Factor-alpha
18.
J Neurosci ; 31(31): 11411-20, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21813701

ABSTRACT

Infantile neuroaxonal dystrophy (INAD) is a fatal neurodegenerative disease characterized by the widespread presence of axonal swellings (spheroids) in the CNS and PNS and is caused by gene abnormality in PLA2G6 [calcium-independent phospholipase A(2)ß (iPLA(2)ß)], which is essential for remodeling of membrane phospholipids. To clarify the pathomechanism of INAD, we pathologically analyzed the spinal cords and sciatic nerves of iPLA(2)ß knock-out (KO) mice, a model of INAD. At 15 weeks (preclinical stage), periodic acid-Schiff (PAS)-positive granules were frequently observed in proximal axons and the perinuclear space of large neurons, and these were strongly positive for a marker of the mitochondrial outer membrane and negative for a marker of the inner membrane. By 100 weeks (late clinical stage), PAS-positive granules and spheroids had increased significantly in the distal parts of axons, and ultrastructural examination revealed that these granules were, in fact, mitochondria with degenerative inner membranes. Collapse of mitochondria in axons was accompanied by focal disappearance of the cytoskeleton. Partial membrane loss at axon terminals was also evident, accompanied by degenerative membranes in the same areas. Imaging mass spectrometry showed a prominent increase of docosahexaenoic acid-containing phosphatidylcholine in the gray matter, suggesting insufficient membrane remodeling in the presence of iPLA(2)ß deficiency. Prominent axonal degeneration in neuroaxonal dystrophy might be explained by the collapse of abnormal mitochondria after axonal transportation. Insufficient remodeling and degeneration of mitochondrial inner membranes and presynaptic membranes appear to be the cause of the neuroaxonal dystrophy in iPLA(2)ß-KO mice.


Subject(s)
Calcium/metabolism , Group VI Phospholipases A2/deficiency , Mitochondria/pathology , Neuroaxonal Dystrophies , Neurodegenerative Diseases/etiology , Presynaptic Terminals/pathology , Age Factors , Aldehydes/metabolism , Animals , Chromatography, Liquid/methods , Disease Models, Animal , Docosahexaenoic Acids/metabolism , Electron Transport Complex IV/metabolism , Female , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/ultrastructure , Models, Biological , Neuroaxonal Dystrophies/complications , Neuroaxonal Dystrophies/genetics , Neuroaxonal Dystrophies/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Spectrometry, Mass, Electrospray Ionization/methods , Spinal Cord/pathology , Spinal Cord/ultrastructure
19.
FASEB J ; 25(12): 4240-52, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21868473

ABSTRACT

Spinal cord injury (SCI) results in permanent loss of motor functions. A significant aspect of the tissue damage and functional loss may be preventable as it occurs, secondary to the trauma. We show that the phospholipase A(2) (PLA(2)) superfamily plays important roles in SCI. PLA(2) enzymes hydrolyze membrane glycerophospholipids to yield a free fatty acid and lysophospholipid. Some free fatty acids (arachidonic acid) give rise to eicosanoids that promote inflammation, while some lysophospholipids (lysophosphatidylcholine) cause demyelination. We show in a mouse model of SCI that two cytosolic forms [calcium-dependent PLA(2) group IVA (cPLA(2) GIVA) and calcium-independent PLA(2) group VIA (iPLA(2) GVIA)], and a secreted form [secreted PLA(2) group IIA (sPLA(2) GIIA)] are up-regulated. Using selective inhibitors and null mice, we show that these PLA(2)s play differing roles. cPLA(2) GIVA mediates protection, whereas sPLA(2) GIIA and, to a lesser extent, iPLA(2) GVIA are detrimental. Furthermore, completely blocking all three PLA(2)s worsens outcome, while the most beneficial effects are seen by partial inhibition of all three. The partial inhibitor enhances expression of cPLA(2) and mediates its beneficial effects via the prostaglandin EP1 receptor. These findings indicate that drugs that inhibit detrimental forms of PLA(2) (sPLA(2) and iPLA2) and up-regulate the protective form (cPLA2) may be useful for the treatment of SCI.


Subject(s)
Phospholipases A2/metabolism , Spinal Cord Injuries/enzymology , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Female , Group II Phospholipases A2/antagonists & inhibitors , Group II Phospholipases A2/deficiency , Group II Phospholipases A2/metabolism , Group IV Phospholipases A2/antagonists & inhibitors , Group IV Phospholipases A2/deficiency , Group IV Phospholipases A2/genetics , Group IV Phospholipases A2/metabolism , Group VI Phospholipases A2/antagonists & inhibitors , Group VI Phospholipases A2/deficiency , Group VI Phospholipases A2/metabolism , Locomotion/drug effects , Locomotion/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Phospholipase A2 Inhibitors , Phospholipases A2/classification , Phospholipases A2/deficiency , Receptor Cross-Talk , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
20.
J Lipid Res ; 51(11): 3166-73, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20686114

ABSTRACT

Ca(2+)-independent phospholipase A(2)ß (iPLA(2)ß) selectively hydrolyzes docosahexaenoic acid (DHA, 22:6n-3) in vitro from phospholipid. Mutations in the PLA2G6 gene encoding this enzyme occur in patients with idiopathic neurodegeneration plus brain iron accumulation and dystonia-parkinsonism without iron accumulation, whereas mice lacking PLA2G6 show neurological dysfunction and neuropathology after 13 months. We hypothesized that brain DHA metabolism and signaling would be reduced in 4-month-old iPLA(2)ß-deficient mice without overt neuropathology. Saline or the cholinergic muscarinic M(1,3,5) receptor agonist arecoline (30 mg/kg) was administered to unanesthetized iPLA(2)ß(-/-), iPLA(2)ß(+/-), and iPLA(2)ß(+/+) mice, and [1-(14)C]DHA was infused intravenously. DHA incorporation coefficients k* and rates J(in), representing DHA metabolism, were determined using quantitative autoradiography in 81 brain regions. iPLA(2)ß(-/-) or iPLA(2)ß(+/-) compared with iPLA(2)ß(+/+) mice showed widespread and significant baseline reductions in k* and J(in) for DHA. Arecoline increased both parameters in brain regions of iPLA(2)ß(+/+) mice but quantitatively less so in iPLA(2)ß(-/-) and iPLA(2)ß(+/-) mice. Consistent with iPLA(2)ß's reported ability to selectively hydrolyze DHA from phospholipid in vitro, iPLA(2)ß deficiency reduces brain DHA metabolism and signaling in vivo at baseline and following M(1,3,5) receptor activation. Positron emission tomography might be used to image disturbed brain DHA metabolism in patients with PLA2G6 mutations.


Subject(s)
Brain/cytology , Brain/metabolism , Docosahexaenoic Acids/metabolism , Group VI Phospholipases A2/deficiency , Molecular Imaging , Signal Transduction , Animals , Arecoline/administration & dosage , Arecoline/pharmacology , Body Weight , Brain/blood supply , Brain/drug effects , Cerebral Arteries/physiology , Docosahexaenoic Acids/blood , Group VI Phospholipases A2/metabolism , Kinetics , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...