Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Physiol Res ; 73(2): 189-203, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38710051

ABSTRACT

This comprehensive review explores the physiological and pathophysiological significance of VPS13A, a protein encoded by the VPS13A gene. The VPS13A gene is associated with Chorea-acanthocytosis (ChAc), a rare hereditary neurodegenerative disorder. The review covers essential aspects, beginning with the genetics of VPS13A, highlighting its role in the pathogenesis of ChAc, and addressing the spectrum of genetic variants involved. It delves into the structure and function of the VPS13A protein, emphasizing its presence in various tissues and its potential involvement in protein trafficking and lipid homeostasis. Molecular functions of VPS13A in the brain tissue and other cell types or tissues with respect to their role in cytoskeletal regulation and autophagy are explored. Finally, it explores the intriguing link between VPS13A mutations, lipid imbalances, and neurodegeneration, shedding light on future research directions. Overall, this review serves as a comprehensive resource for understanding the pivotal role of VPS13A in health and disease, particularly in the context of ChAc. Key words: Chorein , Tumor, Actin, Microfilament, Gene expression, Chorea-acanthocytosis.


Subject(s)
Neuroacanthocytosis , Vesicular Transport Proteins , Humans , Animals , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Neuroacanthocytosis/metabolism , Neuroacanthocytosis/genetics , Neuroacanthocytosis/physiopathology , Neuroacanthocytosis/pathology , Mutation , Lipid Metabolism/physiology , Lipid Metabolism/genetics
2.
Mov Disord ; 38(8): 1535-1541, 2023 08.
Article in English | MEDLINE | ID: mdl-37307400

ABSTRACT

BACKGROUND: Chorea-acanthocytosis (ChAc) is associated with mutations of VPS13A, which encodes for chorein, a protein implicated in lipid transport at intracellular membrane contact sites. OBJECTIVES: The goal of this study was to establish the lipidomic profile of patients with ChAc. METHODS: We analyzed 593 lipid species in the caudate nucleus (CN), putamen, and dorsolateral prefrontal cortex (DLPFC) from postmortem tissues of four patients with ChAc and six patients without ChAc. RESULTS: We found increased levels of bis(monoacylglycerol)phosphate, sulfatide, lysophosphatidylserine, and phosphatidylcholine ether in the CN and putamen, but not in the DLPFC, of patients with ChAc. Phosphatidylserine and monoacylglycerol were increased in the CN and N-acyl phosphatidylserine in the putamen. N-acyl serine was decreased in the CN and DLPFC, whereas lysophosphatidylinositol was decreased in the DLPFC. CONCLUSIONS: We present the first evidence of altered sphingolipid and phospholipid levels in the brains of patients with ChAc. Our observations are congruent with recent findings in cellular and animal models, and implicate defects of lipid processing in VPS13A disease pathophysiology. © 2023 International Parkinson and Movement Disorder Society. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.


Subject(s)
Neuroacanthocytosis , Animals , Humans , Neuroacanthocytosis/genetics , Neuroacanthocytosis/metabolism , Phospholipids/metabolism , Phosphatidylserines/metabolism , Vesicular Transport Proteins/genetics , Brain/metabolism
3.
J Cell Mol Med ; 27(11): 1557-1564, 2023 06.
Article in English | MEDLINE | ID: mdl-37163371

ABSTRACT

VPS13A is a lipid transfer protein localized at different membrane contact sites between organelles, and mutations in the corresponding gene produce a rare neurodegenerative disease called chorea-acanthocytosis (ChAc). Previous studies showed that VPS13A depletion in HeLa cells results in an accumulation of endosomal and lysosomal markers, suggesting a defect in lysosomal degradation capacity leading to partial autophagic dysfunction. Our goal was to determine whether compounds that modulate the endo-lysosomal pathway could be beneficial in the treatment of ChAc. To test this hypothesis, we first generated a KO model using CRISPR/Cas9 to study the consequences of the absence of VPS13A in HeLa cells. We found that inactivation of VPS13A impairs cell growth, which precludes the use of isolated clones due to the undesirable selection of edited clones with residual protein expression. Therefore, we optimized the use of pool cells obtained shortly after transfection with CRISPR/Cas9 components. These cells are a mixture of wild-type and edited cells that allow a comparative analysis of phenotypes and avoids the selection of clones with residual level of VPS13A expression after long-term growth. Consistent with previous observations by siRNA inactivation, VPS13A inactivation by CRISPR/Cas9 resulted in accumulation of the endo-lysosomal markers RAB7A and LAMP1. Notably, we observed that rapamycin partially suppressed the difference in lysosome accumulation between VPS13A KO and WT cells, suggesting that modulation of the autophagic and lysosomal pathway could be a therapeutic target in the treatment of ChAc.


Subject(s)
Neuroacanthocytosis , Neurodegenerative Diseases , Humans , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , CRISPR-Cas Systems/genetics , HeLa Cells , Sirolimus/pharmacology , Neurodegenerative Diseases/metabolism , Lysosomes/metabolism , Neuroacanthocytosis/genetics , Neuroacanthocytosis/metabolism
4.
J Cell Sci ; 135(17)2022 09 01.
Article in English | MEDLINE | ID: mdl-35950506

ABSTRACT

VPS13 family proteins form conduits between the membranes of different organelles through which lipids are transferred. In humans, there are four VPS13 paralogs, and mutations in the genes encoding each of them are associated with different inherited disorders. VPS13 proteins contain multiple conserved domains. The Vps13 adaptor-binding (VAB) domain binds to adaptor proteins that recruit VPS13 to specific membrane contact sites. This work demonstrates the importance of a different domain in VPS13A function. The pleckstrin homology (PH) domain at the C-terminal region of VPS13A is required to form a complex with the XK scramblase and for the co-localization of VPS13A with XK within the cell. Alphafold modeling was used to predict an interaction surface between VPS13A and XK. Mutations in this region disrupt both complex formation and co-localization of the two proteins. Mutant VPS13A alleles found in patients with VPS13A disease truncate the PH domain. The phenotypic similarities between VPS13A disease and McLeod syndrome caused by mutations in VPS13A and XK, respectively, argue that loss of the VPS13A-XK complex is the basis of both diseases.


Subject(s)
Neuroacanthocytosis , Vesicular Transport Proteins , Humans , Mitochondrial Membranes/metabolism , Mutation/genetics , Neuroacanthocytosis/complications , Neuroacanthocytosis/genetics , Neuroacanthocytosis/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
5.
Proc Natl Acad Sci U S A ; 119(35): e2205425119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994651

ABSTRACT

Chorea-acanthocytosis (ChAc) and McLeod syndrome are diseases with shared clinical manifestations caused by mutations in VPS13A and XK, respectively. Key features of these conditions are the degeneration of caudate neurons and the presence of abnormally shaped erythrocytes. XK belongs to a family of plasma membrane (PM) lipid scramblases whose action results in exposure of PtdSer at the cell surface. VPS13A is an endoplasmic reticulum (ER)-anchored lipid transfer protein with a putative role in the transport of lipids at contacts of the ER with other membranes. Recently VPS13A and XK were reported to interact by still unknown mechanisms. So far, however, there is no evidence for a colocalization of the two proteins at contacts of the ER with the PM, where XK resides, as VPS13A was shown to be localized at contacts between the ER and either mitochondria or lipid droplets. Here we show that VPS13A can also localize at ER-PM contacts via the binding of its PH domain to a cytosolic loop of XK, that such interaction is regulated by an intramolecular interaction within XK, and that both VPS13A and XK are highly expressed in the caudate neurons. Binding of the PH domain of VPS13A to XK is competitive with its binding to intracellular membranes that mediate other tethering functions of VPS13A. Our findings support a model according to which VPS13A-dependent lipid transfer between the ER and the PM is coupled to lipid scrambling within the PM. They raise the possibility that defective cell surface exposure of PtdSer may be responsible for neurodegeneration.


Subject(s)
Carrier Proteins , Cell Membrane , Lipids , Vesicular Transport Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , Humans , Neuroacanthocytosis/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
6.
Lab Med ; 53(4): 433-435, 2022 Jul 04.
Article in English | MEDLINE | ID: mdl-35075478

ABSTRACT

Chorea-acanthocytosis (ChAc) is a rare autosomal recessive neurodegenerative disorder caused by pathogenic variants of the vacuolar protein sorting 13A (VPS13A). Only a few patients with ChAc have been reported to date, and the variant spectrum of VPS13A has not been completely elucidated. We describe the case of a 36-year-old woman who had been experiencing orofacial dyskinesia since age 30 years. In a genetic study using next-generation sequencing, 2 variants of VPS13A, the nonsense variant c.4411C>T (p.Arg1471Ter) and the splicing variant c.145-2A>T, were identified. The splicing variant c.145-2A>T was newly classified as a pathogenic variant through a literature review. Consequently, the patient was diagnosed with ChAc based on the typical clinical manifestations, laboratory findings, and imaging results.


Subject(s)
Neuroacanthocytosis , Adult , Female , Humans , Neuroacanthocytosis/diagnosis , Neuroacanthocytosis/genetics , Neuroacanthocytosis/metabolism , Protein Transport , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
7.
Genes (Basel) ; 12(3)2021 02 26.
Article in English | MEDLINE | ID: mdl-33652783

ABSTRACT

Neuroacanthocytosis (NA) syndromes are a group of genetically defined diseases characterized by the association of red blood cell acanthocytosis, progressive degeneration of the basal ganglia and neuromuscular features with characteristic persistent hyperCKemia. The main NA syndromes include autosomal recessive chorea-acanthocytosis (ChAc) and X-linked McLeod syndrome (MLS). A series of Italian patients selected through a multicenter study for these specific neurological phenotypes underwent DNA sequencing of the VPS13A and XK genes to search for causative mutations. Where it has been possible, muscle biopsies were obtained and thoroughly investigated with histochemical assays. A total of nine patients from five different families were diagnosed with ChAC and had mostly biallelic changes in the VPS13A gene (three nonsense, two frameshift, three splicing), while three patients from a single X-linked family were diagnosed with McLeod syndrome and had a deletion in the XK gene. Despite a very low incidence (only one thousand cases of ChAc and a few hundred MLS cases reported worldwide), none of the 8 VPS13A variants identified in our patients is shared by two families, suggesting the high genetic variability of ChAc in the Italian population. In our series, in line with epidemiological data, McLeod syndrome occurs less frequently than ChAc, although it can be easily suspected because of its X-linked mode of inheritance. Finally, histochemical studies strongly suggest that muscle pathology is not simply secondary to the axonal neuropathy, frequently seen in these patients, but primary myopathic alterations can be detected in both NA syndromes.


Subject(s)
Muscle, Skeletal , Mutation , Vesicular Transport Proteins , Adult , Child , Cohort Studies , Erythrocytes/metabolism , Erythrocytes/pathology , Female , Humans , Italy , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Muscular Diseases/metabolism , Muscular Diseases/pathology , Neuroacanthocytosis/genetics , Neuroacanthocytosis/metabolism , Neuroacanthocytosis/pathology , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
8.
Mol Biol Cell ; 31(22): 2425-2436, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32845802

ABSTRACT

Vps13 is a highly conserved lipid transfer protein found at multiple interorganelle membrane contact sites where it mediates distinct processes. In yeast, recruitment of Vps13 to different contact sites occurs via various partner proteins. In humans, four VPS13 family members, A-D, are associated with different diseases. In particular, vps13A mutants result in the neurodegenerative disorder Chorea-Acanthocytosis (ChAc). ChAc phenotypes resemble those of McLeod Syndrome, caused by mutations in the XK gene, suggesting that XK could be a partner protein for VPS13A. XK does, in fact, exhibit hallmarks of a VPS13A partner: it forms a complex with VPS13A in human cells and, when overexpressed, relocalizes VPS13A from lipid droplets to subdomains of the endoplasmic reticulum. Introduction of two different ChAc disease-linked missense mutations into VPS13A prevents this XK-induced relocalization. These results suggest that dysregulation of a VPS13A-XK complex is the common basis for ChAc and McLeod Syndrome.


Subject(s)
Amino Acid Transport Systems, Neutral/metabolism , Neuroacanthocytosis/metabolism , Vesicular Transport Proteins/metabolism , Amino Acid Transport Systems, Neutral/genetics , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , HEK293 Cells , HeLa Cells , Humans , Lipid Droplets/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Neuroacanthocytosis/genetics , Vesicular Transport Proteins/genetics
9.
Sci Rep ; 10(1): 8391, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32439941

ABSTRACT

Loss of function mutations of the chorein-encoding gene VPS13A lead to chorea-acanthocytosis (ChAc), a neurodegenerative disorder with accelerated suicidal neuronal cell death, which could be reversed by lithium. Chorein upregulates the serum and glucocorticoid inducible kinase SGK1. Targets of SGK1 include the Na+/K+-ATPase, a pump required for cell survival. To explore whether chorein-deficiency affects Na+/K+ pump capacity, cortical neurons were differentiated from iPSCs generated from fibroblasts of ChAc patients and healthy volunteers. Na+/K+ pump capacity was estimated from K+-induced whole cell outward current (pump capacity). As a result, the pump capacity was completely abolished in the presence of Na+/K+ pump-inhibitor ouabain (100 µM), was significantly smaller in ChAc neurons than in control neurons, and was significantly increased in ChAc neurons by lithium treatment (24 hours 2 mM). The effect of lithium was reversed by SGK1-inhibitor GSK650394 (24 h 10 µM). Transmembrane potential (Vm) was significantly less negative in ChAc neurons than in control neurons, and was significantly increased in ChAc neurons by lithium treatment (2 mM, 24 hours). The effect of lithium on Vm was virtually abrogated by ouabain. Na+/K+ α1-subunit transcript levels and protein abundance were significantly lower in ChAc neurons than in control neurons, an effect reversed by lithium treatment (2 mM, 24 hours). In conclusion, consequences of chorein deficiency in ChAc include impaired Na+/K+ pump capacity.


Subject(s)
Cell Membrane/pathology , Neuroacanthocytosis/metabolism , Neuroacanthocytosis/pathology , Neurons/pathology , Sodium-Potassium-Exchanging ATPase/metabolism , Benzoates/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Case-Control Studies , Cell Differentiation , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Humans , Immediate-Early Proteins/antagonists & inhibitors , Induced Pluripotent Stem Cells/cytology , Lithium/pharmacology , Membrane Potentials/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/genetics , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
10.
Biochim Biophys Acta Biomembr ; 1862(9): 183349, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32407779

ABSTRACT

Chorein is a protein of the Vps13 family, and defects in this protein cause the rare neurodegenerative disorder chorea-acanthocytosis (ChAc). Chorein is involved in the actin cytoskeleton organization, calcium ion flux, neuronal cell excitability, exocytosis and autophagy. The function of this protein is poorly understood, and obtaining this knowledge is a key to finding a cure for ChAc. Chorein, as well as the Vps13 protein from yeast, contains the APT1 domain. Our previous research has shown that the APT1 domain from yeast Vps13 (yAPT1v) binds phosphatidylinositol 3-phosphate (PI3P) in vitro. In this study, we showed that although the APT1 domain from chorein (hAPT1) binds to PI3P it could not functionally replace yAPT1v. The hAPT1 domain binds, in addition to PI3P, to phosphatidylinositol 5-phosphate (PI5P). The binding of hAPT1 to PI3P, unlike the binding of yAPT1v to PI3P, is regulated by the bivalent ions, calcium and magnesium. Regulation of PI3P binding via calcium is also observed for the APT1 domain of yeast autophagy protein Atg2. The substitution I2771R, found in chorein of patient suffering from ChAc, reduces the binding of the hAPT1 domain to PI3P and PI5P. These results suggest that the ability of APT1 domains to bind phosphoinositides is regulated differently in yeast and human protein and that this regulation is important for chorein function.


Subject(s)
Neuroacanthocytosis/genetics , Saccharomyces cerevisiae Proteins/genetics , Thiolester Hydrolases/genetics , Vesicular Transport Proteins/genetics , Autophagy/genetics , Autophagy-Related Proteins/genetics , Calcium/chemistry , Humans , Ions/chemistry , Magnesium/chemistry , Mutation/genetics , Neuroacanthocytosis/metabolism , Neuroacanthocytosis/pathology , Neurons/metabolism , Neurons/pathology , Phosphatidylinositol Phosphates/genetics , Protein Binding/genetics , Protein Domains/genetics , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/chemistry , Thiolester Hydrolases/chemistry , Vesicular Transport Proteins/chemistry
11.
Dis Model Mech ; 12(1)2019 01 28.
Article in English | MEDLINE | ID: mdl-30635263

ABSTRACT

Chorea-acanthocytosis (ChAc) is a rare neurodegenerative disease associated with mutations in the human VPS13A gene. The mechanism of ChAc pathogenesis is unclear. A simple yeast model was used to investigate the function of the single yeast VSP13 orthologue, Vps13. Vps13, like human VPS13A, is involved in vesicular protein transport, actin cytoskeleton organisation and phospholipid metabolism. A newly identified phenotype of the vps13Δ mutant, sodium dodecyl sulphate (SDS) hypersensitivity, was used to screen a yeast genomic library for multicopy suppressors. A fragment of the MYO3 gene, encoding Myo3-N (the N-terminal part of myosin, a protein involved in the actin cytoskeleton and in endocytosis), was isolated. Myo3-N protein contains a motor head domain and a linker. The linker contains IQ motifs that mediate the binding of calmodulin, a negative regulator of myosin function. Amino acid substitutions that disrupt the interaction of Myo3-N with calmodulin resulted in the loss of vps13Δ suppression. Production of Myo3-N downregulated the activity of calcineurin, a protein phosphatase regulated by calmodulin, and alleviated some defects in early endocytosis events. Importantly, ethylene glycol tetraacetic acid (EGTA), which sequesters calcium and thus downregulates calmodulin and calcineurin, was a potent suppressor of vps13Δ. We propose that Myo3-N acts by sequestering calmodulin, downregulating calcineurin and increasing activity of Myo3, which is involved in endocytosis and, together with Osh2/3 proteins, functions in endoplasmic reticulum-plasma membrane contact sites. These results show that defects associated with vps13Δ could be overcome, and point to a functional connection between Vps13 and calcium signalling as a possible target for chemical intervention in ChAc. Yeast ChAc models may uncover the underlying pathological mechanisms, and may also serve as a platform for drug testing.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Calcium Signaling , Calcium/metabolism , Calmodulin/metabolism , Models, Biological , Myosins/metabolism , Neuroacanthocytosis/drug therapy , Neuroacanthocytosis/metabolism , Saccharomyces cerevisiae/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Alleles , Amino Acid Substitution , Calcineurin/metabolism , Calcium Signaling/drug effects , Canavanine/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Endocytosis/drug effects , Genes, Suppressor , Mutation/genetics , Protein Domains , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/metabolism , Sodium Dodecyl Sulfate , Transcription, Genetic/drug effects , Vacuoles/metabolism
12.
Mol Cell Neurosci ; 92: 137-148, 2018 10.
Article in English | MEDLINE | ID: mdl-30081151

ABSTRACT

Mutations in the VPS13A gene leading to depletion of chorein protein are causative for Chorea Acanthocytosis (ChAc), a rare devastating disease, which is characterized by neurodegeneration mainly affecting the basal ganglia as well as deformation of erythrocytes. Studies on patient blood samples highlighted a dysregulation of Actin cytoskeleton caused by downregulation of the PI3K pathway and hyper-activation of Lyn-kinase, but to what extent these mechanisms are present and relevant in the affected neurons remains elusive. We studied the effects of the absence of chorein protein on the morphology and trafficking of lysosomal and mitochondrial compartments in ChAc patient-specific induced pluripotent stem cell-derived medium spiny neurons (MSNs). Numbers of both organelle types were reduced in ChAc MSNs. Mitochondrial length was shortened and their membrane potential showed significant hyperpolarization. In contrast to previous studies, showing Lyn kinase dependency of ChAc-associated pathological events in erythrocytes, pharmacological studies demonstrate that the impairment of mitochondria and lysosomes are independent of Lyn kinase activity. These data suggest that impairment in mitochondrial and lysosomal morphologies in MSNs is not mediated by a dysregulation of Lyn kinase and thus the pathological pathways in ChAc might be - at least in part - cell-type specific.


Subject(s)
Lysosomes/metabolism , Mitochondria/metabolism , Neuroacanthocytosis/metabolism , src-Family Kinases/metabolism , Adult , Cells, Cultured , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Lysosomes/pathology , Male , Membrane Potential, Mitochondrial , Middle Aged , Mitochondria/pathology , Neuroacanthocytosis/genetics , Neuroacanthocytosis/pathology , Signal Transduction , Vesicular Transport Proteins/genetics
13.
Neurosignals ; 25(1): 117-126, 2017.
Article in English | MEDLINE | ID: mdl-29179176

ABSTRACT

Chorea-acanthocytosis (ChAc), a neurodegenerative disease, results from loss-of-function-mutations of the chorein-encoding gene VPS13A. Affected patients suffer from a progressive movement disorder including chorea, parkinsonism, dystonia, tongue protrusion, dysarthria, dysphagia, tongue and lip biting, gait impairment, progressive distal muscle wasting, weakness, epileptic seizures, cognitive impairment, and behavioral changes. Those pathologies may be paralleled by erythrocyte acanthocytosis. Chorein supports activation of phosphoinositide-3-kinase (PI3K)-p85-subunit with subsequent up-regulation of ras-related C3 botulinum toxin substrate 1 (Rac1) activity, p21 protein-activated kinase 1 (PAK1) phosphorylation, and activation of several tyrosine kinases. Chorein sensitive PI3K signaling further leads to stimulation of the serum and glucocorticoid inducible kinase SGK1, which in turn upregulates ORAI1, a Ca2+-channel accomplishing store operated Ca2+-entry (SOCE). The signaling participates in the regulation of cytoskeletal architecture on the one side and cell survival on the other. Compromised cytoskeletal architecture has been shown in chorein deficient erythrocytes, fibroblasts and endothelial cells. Impaired degranulation was observed in chorein deficient PC12 cells and in platelets from ChAc patients. Similarly, decreased ORAI1 expression and SOCE as well as compromised cell survival were seen in fibroblasts and neurons isolated from ChAc patients. ORAI1 expression, SOCE and cell survival can be restored by lithium treatment, an effect disrupted by pharmacological inhibition of SGK1 or ORAI1. Chorein, SGK1, ORAI1 and SOCE further confer survival of tumor cells. In conclusion, much has been learned about the function of chorein and the molecular pathophysiology of chorea-acanthocytosis. Most importantly, a treatment halting or delaying the clinical course of this devastating disease may become available. A controlled clinical study is warranted, in order to explore whether the in vitro observations indeed reflect the in vivo pathology of the disease.


Subject(s)
Erythrocytes/metabolism , Neuroacanthocytosis/metabolism , Neurons/metabolism , Vesicular Transport Proteins/metabolism , Animals , Autophagy/physiology , Cytoskeleton/metabolism , Humans
14.
Cell Physiol Biochem ; 42(5): 2066-2077, 2017.
Article in English | MEDLINE | ID: mdl-28803243

ABSTRACT

BACKGROUND: The widely expressed protein chorein fosters activation of the phosphoinositide 3 kinase (PI3K) pathway thus supporting cell survival. Loss of function mutations of the chorein encoding gene VPS13A (vacuolar protein sorting-associated protein 13A) causes chorea-acanthocytosis (ChAc), a neurodegenerative disorder paralleled by deformations of erythrocytes. In mice, genetic knockout of chorein leads to enhanced neuronal apoptosis. PI3K dependent signalling upregulates Orai1, a pore forming channel protein accomplishing store operated Ca2+ entry (SOCE). Increased Orai1 expression and SOCE have been shown to confer survival of tumor cells. SOCE could be up-regulated by lithium. The present study explored, whether SOCE and/or apoptosis are altered in ChAc fibroblasts and could be modified by lithium treatment. METHODS: Fibroblasts were isolated from ChAc patients and age-matched healthy volunteers. Cytosolic Ca2+ activity ([Ca2+]i) was estimated from Fura-2-fluorescence, SOCE from increase of [Ca2+]i following Ca2+ re-addition after Ca2+-store depletion with sarcoendoplasmatic Ca2+-ATPase (SERCA) inhibitor thapsigargin (1 µM), and apoptosis from annexin-V/propidium iodide staining quantified in flow cytometry. RESULTS: SOCE was significantly smaller in ChAc fibroblasts than in control fibroblasts. Lithium (2 mM, 24 hours) significantly increased and Orai1 blocker 2-Aminoethoxydiphenyl Borate (2-APB, 50 µM, 24 hours) significantly decreased SOCE. Annexin-V-binding and propidium iodide staining were significantly higher in ChAc fibroblasts than in control fibroblasts. In ChAc fibroblasts annexin-V-binding and propidium iodide staining were significantly decreased by lithium treatment, significantly increased by 2-APB and virtually lithium insensitive in the presence of 2-APB. CONCLUSIONS: In ChAc fibroblasts, downregulation of SOCE contributes to enhanced susceptibility to apoptosis. Both, decreased SOCE and enhanced apoptosis of ChAc fibroblasts can be reversed by lithium treatment.


Subject(s)
Calcium Release Activated Calcium Channels/metabolism , Fibroblasts/drug effects , Lithium/pharmacology , Neuroacanthocytosis/pathology , Apoptosis/drug effects , Boron Compounds/pharmacology , Calcium/metabolism , Calcium Release Activated Calcium Channels/antagonists & inhibitors , Calcium-Transporting ATPases/metabolism , Case-Control Studies , Cell Survival/drug effects , Cells, Cultured , Down-Regulation/drug effects , Fibroblasts/cytology , Fibroblasts/metabolism , Fura-2/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Microscopy, Fluorescence , Neuroacanthocytosis/metabolism
15.
Traffic ; 18(11): 711-719, 2017 11.
Article in English | MEDLINE | ID: mdl-28846184

ABSTRACT

Human Vps13 proteins are associated with several diseases, including the neurodegenerative disorder Chorea-acanthocytosis (ChAc), yet the biology of these proteins is still poorly understood. Studies in Saccharomyces cerevisiae, Dictyostelium discoideum, Tetrahymena thermophila and Drosophila melanogaster point to the involvement of Vps13 in cytoskeleton organization, vesicular trafficking, autophagy, phagocytosis, endocytosis, proteostasis, sporulation and mitochondrial functioning. Recent findings show that yeast Vps13 binds to phosphatidylinositol lipids via 4 different regions and functions at membrane contact sites, enlarging the list of Vps13 functions. This review describes the great potential of simple eukaryotes to decipher disease mechanisms in higher organisms and highlights novel insights into the pathological role of Vps13 towards ChAc.


Subject(s)
Neuroacanthocytosis/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Vesicular Transport Proteins/metabolism , Animals , Dictyostelium/metabolism , Drosophila melanogaster/metabolism , Humans , Mutation , Neuroacanthocytosis/genetics , Neuroacanthocytosis/pathology , Protein Transport , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Species Specificity , Vesicular Transport Proteins/genetics
16.
Sci Rep ; 7(1): 6457, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28743945

ABSTRACT

Chorea-Acanthocytosis (ChAc), a neurodegenerative disorder, results from loss-of-function-mutations of chorein-encoding gene VPS13A. In tumour cells chorein up-regulates ORAI1, a Ca2+-channel accomplishing store operated Ca2+-entry (SOCE) upon stimulation by STIM1. Furthermore SOCE could be up-regulated by lithium. The present study explored whether SOCE impacts on neuron apoptosis. Cortical neurons were differentiated from induced pluripotent stem cells generated from fibroblasts of ChAc patients and healthy volunteers. ORAI1 and STIM1 transcript levels and protein abundance were estimated from qRT-PCR and Western blotting, respectively, cytosolic Ca2+-activity ([Ca2+]i) from Fura-2-fluorescence, as well as apoptosis from annexin-V-binding and propidium-iodide uptake determined by flow cytometry. As a result, ORAI1 and STIM1 transcript levels and protein abundance and SOCE were significantly smaller and the percentage apoptotic cells significantly higher in ChAc neurons than in control neurons. Lithium treatment (2 mM, 24 hours) increased significantly ORAI1 and STIM1 transcript levels and protein abundance, an effect reversed by inhibition of Serum & Glucocorticoid inducible Kinase 1. ORAI1 blocker 2-APB (50 µM, 24 hours) significantly decreased SOCE, markedly increased apoptosis and abrogated the anti-apoptotic effect of lithium. In conclusion, enhanced neuronal apoptosis in ChAc at least partially results from decreased ORAI1 expression and SOCE, which could be reversed by lithium treatment.


Subject(s)
Calcium/metabolism , Lithium/pharmacology , Neuroacanthocytosis/pathology , Neurons/pathology , ORAI1 Protein/metabolism , Apoptosis/drug effects , Benzoates/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Death , Cell Differentiation , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Healthy Volunteers , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neuroacanthocytosis/metabolism , Neurons/drug effects , Neurons/metabolism , ORAI1 Protein/genetics , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism
17.
Neurosci Lett ; 654: 107-110, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28645789

ABSTRACT

Chorea-acanthocytosis (Ch-Ac) is an autosomal recessive neurodegenerative disorder characterized by adult-onset chorea, acanthocytes in the peripheral blood, and Huntington's disease-like neuropsychiatric symptoms. Animal studies have shown mutation-related dysregulated cortical gamma-aminobutyric acid (GABA)ergic inhibitory networks in its pathophysiology. Herein we found that in patients with Ch-Ac there is a striking alteration of intracortical inhibitory circuits detected by using paired pulse transcranial magnetic stimulation protocols. Our findings show in vivo the functional disruption of GABA(A)-mediated networks in humans with Ch-Ac supporting the existing data in mice models with this condition.


Subject(s)
Motor Cortex/metabolism , Neural Pathways/metabolism , Neuroacanthocytosis/metabolism , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/metabolism , Humans , Male , Middle Aged , Motor Cortex/physiopathology , Neural Pathways/physiopathology , Neuroacanthocytosis/physiopathology , Synaptic Transmission/physiology , Transcranial Magnetic Stimulation
19.
J Neurosci ; 36(47): 12027-12043, 2016 11 23.
Article in English | MEDLINE | ID: mdl-27881786

ABSTRACT

Chorea-acanthocytosis (ChAc) is a fatal neurological disorder characterized by red blood cell acanthocytes and striatal neurodegeneration. Recently, severe cell membrane disturbances based on depolymerized cortical actin and an elevated Lyn kinase activity in erythrocytes from ChAc patients were identified. How this contributes to the mechanism of neurodegeneration is still unknown. To gain insight into the pathophysiology, we established a ChAc patient-derived induced pluripotent stem cell model and an efficient differentiation protocol providing a large population of human striatal medium spiny neurons (MSNs), the main target of neurodegeneration in ChAc. Patient-derived MSNs displayed enhanced neurite outgrowth and ramification, whereas synaptic density was similar to controls. Electrophysiological analysis revealed a pathologically elevated synaptic activity in ChAc MSNs. Treatment with the F-actin stabilizer phallacidin or the Src kinase inhibitor PP2 resulted in the significant reduction of disinhibited synaptic currents to healthy control levels, suggesting a Src kinase- and actin-dependent mechanism. This was underlined by increased G/F-actin ratios and elevated Lyn kinase activity in patient-derived MSNs. These data indicate that F-actin stabilization and Src kinase inhibition represent potential therapeutic targets in ChAc that may restore neuronal function. SIGNIFICANCE STATEMENT: Chorea-acanthocytosis (ChAc) is a fatal neurodegenerative disease without a known cure. To gain pathophysiological insight, we newly established a human in vitro model using skin biopsies from ChAc patients to generate disease-specific induced pluripotent stem cells (iPSCs) and developed an efficient iPSC differentiation protocol providing striatal medium spiny neurons. Using patch-clamp electrophysiology, we detected a pathologically enhanced synaptic activity in ChAc neurons. Healthy control levels of synaptic activity could be restored by treatment of ChAc neurons with the F-actin stabilizer phallacidin and the Src kinase inhibitor PP2. Because Src kinases are involved in bridging the membrane to the actin cytoskeleton by membrane protein phosphorylation, our data suggest an actin-dependent mechanism of this dysfunctional phenotype and potential treatment targets in ChAc.


Subject(s)
Actins/metabolism , Corpus Striatum/pathology , GABAergic Neurons/pathology , Induced Pluripotent Stem Cells/pathology , Neuroacanthocytosis/metabolism , Neuroacanthocytosis/pathology , src-Family Kinases/metabolism , Adult , Cell Differentiation , Cells, Cultured , Corpus Striatum/metabolism , Female , GABAergic Neurons/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Neurons , Synaptic Transmission , src-Family Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...