Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Country/Region as subject
Publication year range
1.
EMBO J ; 40(8): e103811, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33644875

ABSTRACT

HSP27 is a human molecular chaperone that forms large, dynamic oligomers and functions in many aspects of cellular homeostasis. Mutations in HSP27 cause Charcot-Marie-Tooth (CMT) disease, the most common inherited disorder of the peripheral nervous system. A particularly severe form of CMT disease is triggered by the P182L mutation in the highly conserved IxI/V motif of the disordered C-terminal region, which interacts weakly with the structured core domain of HSP27. Here, we observed that the P182L mutation disrupts the chaperone activity and significantly increases the size of HSP27 oligomers formed in vivo, including in motor neurons differentiated from CMT patient-derived stem cells. Using NMR spectroscopy, we determined that the P182L mutation decreases the affinity of the HSP27 IxI/V motif for its own core domain, leaving this binding site more accessible for other IxI/V-containing proteins. We identified multiple IxI/V-bearing proteins that bind with higher affinity to the P182L variant due to the increased availability of the IxI/V-binding site. Our results provide a mechanistic basis for the impact of the P182L mutation on HSP27 and suggest that the IxI/V motif plays an important, regulatory role in modulating protein-protein interactions.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Heat-Shock Proteins/chemistry , Molecular Chaperones/chemistry , Adult , Binding Sites , Cells, Cultured , HeLa Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Molecular Dynamics Simulation , Motor Neurons/cytology , Motor Neurons/metabolism , Mutation, Missense , Protein Binding , Protein Multimerization
2.
Brain ; 146(7): 2885-2896, 2023 07 03.
Article in English | MEDLINE | ID: mdl-36511878

ABSTRACT

Charcot-Marie-Tooth disease is the most common inherited disorder of the PNS. CMT1A accounts for 40-50% of all cases and is caused by a duplication of the PMP22 gene on chromosome 17, leading to dysmyelination in the PNS. Patient-derived models to study such myelination defects are lacking as the in vitro generation of human myelinating Schwann cells has proved to be particularly challenging. Here, we present an induced pluripotent stem cell-derived organoid culture, containing various cell types of the PNS, including myelinating human Schwann cells, which mimics the human PNS. Single-cell analysis confirmed the PNS-like cellular composition and provides insight into the developmental trajectory. We used this organoid model to study disease signatures of CMT1A, revealing early ultrastructural myelin alterations, including increased myelin periodic line distance and hypermyelination of small axons. Furthermore, we observed the presence of onion-bulb-like formations in a later developmental stage. These hallmarks were not present in the CMT1A-corrected isogenic line or in a CMT2A iPSC line, supporting the notion that these alterations are specific to CMT1A. Downregulation of PMP22 expression using short-hairpin RNAs or a combinatorial drug consisting of baclofen, naltrexone hydrochloride and D-sorbitol was able to ameliorate the myelin defects in CMT1A-organoids. In summary, this self-organizing organoid model can capture biologically meaningful features of the disease and capture the physiological complexity, forms an excellent model for studying demyelinating diseases and supports the therapeutic approach of reducing PMP22 expression.


Subject(s)
Charcot-Marie-Tooth Disease , Induced Pluripotent Stem Cells , Humans , Myelin Sheath/metabolism , Induced Pluripotent Stem Cells/metabolism , Down-Regulation , Myelin Proteins/genetics , Myelin Proteins/metabolism , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Organoids/metabolism , Schwann Cells
3.
Genet Med ; 24(7): 1583-1591, 2022 07.
Article in English | MEDLINE | ID: mdl-35499524

ABSTRACT

PURPOSE: CTR9 is a subunit of the PAF1 complex (PAF1C) that plays a crucial role in transcription regulation by binding CTR9 to RNA polymerase II. It is involved in transcription-coupled histone modification through promoting H3K4 and H3K36 methylation. We describe the clinical and molecular studies in 13 probands, harboring likely pathogenic CTR9 missense variants, collected through GeneMatcher. METHODS: Exome sequencing was performed in all individuals. CTR9 variants were assessed through 3-dimensional modeling of the activated human transcription complex Pol II-DSIF-PAF-SPT6 and the PAF1/CTR9 complex. H3K4/H3K36 methylation analysis, mitophagy assessment based on tetramethylrhodamine ethyl ester perchlorate immunofluorescence, and RNA-sequencing in skin fibroblasts from 4 patients was performed. RESULTS: Common clinical findings were variable degrees of intellectual disability, hypotonia, joint hyperlaxity, speech delay, coordination problems, tremor, and autism spectrum disorder. Mild dysmorphism and cardiac anomalies were less frequent. For 11 CTR9 variants, de novo occurrence was shown. Three-dimensional modeling predicted a likely disruptive effect of the variants on local CTR9 structure and protein interaction. Additional studies in fibroblasts did not unveil the downstream functional consequences of the identified variants. CONCLUSION: We describe a neurodevelopmental disorder caused by (mainly) de novo variants in CTR9, likely affecting PAF1C function.


Subject(s)
Autism Spectrum Disorder , Intellectual Disability , Neurodevelopmental Disorders , Phosphoproteins , Transcription Factors , Gene Expression Regulation , Heterozygote , Humans , Intellectual Disability/genetics , Neurodevelopmental Disorders/genetics , Phosphoproteins/genetics , Transcription Factors/genetics
4.
Brain ; 144(8): 2471-2485, 2021 09 04.
Article in English | MEDLINE | ID: mdl-34128983

ABSTRACT

Axonal Charcot-Marie-Tooth neuropathies (CMT type 2) are caused by inherited mutations in various genes functioning in different pathways. The types of genes and multiplicity of mutations reflect the clinical and genetic heterogeneity in CMT2 disease, which complicates its diagnosis and has inhibited the development of therapies. Here, we used CMT2 patient-derived pluripotent stem cells (iPSCs) to identify common hallmarks of axonal degeneration shared by different CMT2 subtypes. We compared the cellular phenotypes of neurons differentiated from CMT2 patient iPSCs with those from healthy controls and a CRISPR/Cas9-corrected isogenic line. Our results demonstrated neurite network alterations along with extracellular electrophysiological abnormalities in the differentiated motor neurons. Progressive deficits in mitochondrial and lysosomal trafficking, as well as in mitochondrial morphology, were observed in all CMT2 patient lines. Differentiation of the same CMT2 iPSC lines into peripheral sensory neurons only gave rise to cellular phenotypes in subtypes with sensory involvement, supporting the notion that some gene mutations predominantly affect motor neurons. We revealed a common mitochondrial dysfunction in CMT2-derived motor neurons, supported by alterations in the expression pattern and oxidative phosphorylation, which could be recapitulated in the sciatic nerve tissue of a symptomatic mouse model. Inhibition of a dual leucine zipper kinase could partially ameliorate the mitochondrial disease phenotypes in CMT2 subtypes. Altogether, our data reveal shared cellular phenotypes across different CMT2 subtypes and suggests that targeting such common pathomechanisms could allow the development of a uniform treatment for CMT2.


Subject(s)
Charcot-Marie-Tooth Disease/metabolism , Mitochondria/metabolism , Motor Neurons/metabolism , Charcot-Marie-Tooth Disease/pathology , Genotype , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mitochondria/pathology , Motor Neurons/pathology , Mutation , Pedigree
5.
Hum Mol Genet ; 28(4): 615-627, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30339187

ABSTRACT

Axonopathies are neurodegenerative disorders caused by axonal degeneration, affecting predominantly the longest neurons. Several of these axonopathies are caused by genetic defects in proteins involved in the shaping and dynamics of the endoplasmic reticulum (ER); however, it is unclear how these defects impinge on neuronal survival. Given its central and widespread position within a cell, the ER is a pivotal player in inter-organelle communication. Here, we demonstrate that defects in the ER fusion protein ATL3, which were identified in patients suffering from hereditary sensory and autonomic neuropathy, result in an increased number of ER-mitochondria contact sites both in HeLa cells and in patient-derived fibroblasts. This increased contact is reflected in higher phospholipid metabolism, upregulated autophagy and augmented Ca2+ crosstalk between both organelles. Moreover, the mitochondria in these cells display lowered motility, and the number of axonal mitochondria in neurons expressing disease-causing mutations in ATL3 is strongly decreased. These results underscore the functional interdependence of subcellular organelles in health and disease and show that disorders caused by ER-shaping defects are more complex than previously assumed.


Subject(s)
Axons/metabolism , Endoplasmic Reticulum/genetics , GTP Phosphohydrolases/genetics , Hereditary Sensory and Autonomic Neuropathies/genetics , Autophagy/genetics , Axons/pathology , Calcium/metabolism , Calcium Signaling/genetics , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , HeLa Cells , Hereditary Sensory and Autonomic Neuropathies/metabolism , Hereditary Sensory and Autonomic Neuropathies/pathology , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mutation , Neurons/metabolism , Neurons/pathology
6.
Brain ; 142(9): 2605-2616, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31332438

ABSTRACT

Distal hereditary motor neuropathies are a rare subgroup of inherited peripheral neuropathies hallmarked by a length-dependent axonal degeneration of lower motor neurons without significant involvement of sensory neurons. We identified patients with heterozygous nonsense mutations in the αII-spectrin gene, SPTAN1, in three separate dominant hereditary motor neuropathy families via next-generation sequencing. Variable penetrance was noted for these mutations in two of three families, and phenotype severity differs greatly between patients. The mutant mRNA containing nonsense mutations is broken down by nonsense-mediated decay and leads to reduced protein levels in patient cells. Previously, dominant-negative αII-spectrin gene mutations were described as causal in a spectrum of epilepsy phenotypes.


Subject(s)
Carrier Proteins/genetics , Codon, Nonsense/genetics , Microfilament Proteins/genetics , Mutation/genetics , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/genetics , Adult , Aged , Aged, 80 and over , Child , Cohort Studies , Female , Humans , Male , Middle Aged , Pedigree , Young Adult
7.
Acta Neuropathol ; 135(1): 131-148, 2018 01.
Article in English | MEDLINE | ID: mdl-28780615

ABSTRACT

Mutations in the small heat shock protein B8 gene (HSPB8/HSP22) have been associated with distal hereditary motor neuropathy, Charcot-Marie-Tooth disease, and recently distal myopathy. It is so far not clear how mutant HSPB8 induces the neuronal and muscular phenotypes and if a common pathogenesis lies behind these diseases. Growing evidence points towards a role of HSPB8 in chaperone-associated autophagy, which has been shown to be a determinant for the clearance of poly-glutamine aggregates in neurodegenerative diseases but also for the maintenance of skeletal muscle myofibrils. To test this hypothesis and better dissect the pathomechanism of mutant HSPB8, we generated a new transgenic mouse model leading to the expression of the mutant protein (knock-in lines) or the loss-of-function (functional knock-out lines) of the endogenous protein Hspb8. While the homozygous knock-in mice developed motor deficits associated with degeneration of peripheral nerves and severe muscle atrophy corroborating patient data, homozygous knock-out mice had locomotor performances equivalent to those of wild-type animals. The distal skeletal muscles of the post-symptomatic homozygous knock-in displayed Z-disk disorganisation, granulofilamentous material accumulation along with Hspb8, αB-crystallin (HSPB5/CRYAB), and desmin aggregates. The presence of the aggregates correlated with reduced markers of effective autophagy. The sciatic nerve of the homozygous knock-in mice was characterized by low autophagy potential in pre-symptomatic and Hspb8 aggregates in post-symptomatic animals. On the other hand, the sciatic nerve of the homozygous knock-out mice presented a normal morphology and their distal muscle displayed accumulation of abnormal mitochondria but intact myofiber and Z-line organisation. Our data, therefore, suggest that toxic gain-of-function of mutant Hspb8 aggregates is a major contributor to the peripheral neuropathy and the myopathy. In addition, mutant Hspb8 induces impairments in autophagy that may aggravate the phenotype.


Subject(s)
Distal Myopathies/metabolism , Gain of Function Mutation , HSP20 Heat-Shock Proteins/genetics , HSP20 Heat-Shock Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myopathies, Structural, Congenital/metabolism , Peripheral Nervous System Diseases/metabolism , Animals , Atrophy/metabolism , Atrophy/pathology , Autophagy/physiology , Disease Models, Animal , Distal Myopathies/pathology , Female , Heat-Shock Proteins , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/pathology , Molecular Chaperones , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myopathies, Structural, Congenital/pathology , Sciatic Nerve/metabolism , Sciatic Nerve/pathology
8.
Brain ; 140(2): 287-305, 2017 02.
Article in English | MEDLINE | ID: mdl-28137957

ABSTRACT

Hereditary spastic paraplegias are heterogeneous neurodegenerative disorders characterized by progressive spasticity of the lower limbs due to degeneration of the corticospinal motor neurons. In a Bulgarian family with three siblings affected by complicated hereditary spastic paraplegia, we performed whole exome sequencing and homozygosity mapping and identified a homozygous p.Thr512Ile (c.1535C > T) mutation in ATP13A2. Molecular defects in this gene have been causally associated with Kufor-Rakeb syndrome (#606693), an autosomal recessive form of juvenile-onset parkinsonism, and neuronal ceroid lipofuscinosis (#606693), a neurodegenerative disorder characterized by the intracellular accumulation of autofluorescent lipopigments. Further analysis of 795 index cases with hereditary spastic paraplegia and related disorders revealed two additional families carrying truncating biallelic mutations in ATP13A2. ATP13A2 is a lysosomal P5-type transport ATPase, the activity of which critically depends on catalytic autophosphorylation. Our biochemical and immunocytochemical experiments in COS-1 and HeLa cells and patient-derived fibroblasts demonstrated that the hereditary spastic paraplegia-associated mutations, similarly to the ones causing Kufor-Rakeb syndrome and neuronal ceroid lipofuscinosis, cause loss of ATP13A2 function due to transcript or protein instability and abnormal intracellular localization of the mutant proteins, ultimately impairing the lysosomal and mitochondrial function. Moreover, we provide the first biochemical evidence that disease-causing mutations can affect the catalytic autophosphorylation activity of ATP13A2. Our study adds complicated hereditary spastic paraplegia (SPG78) to the clinical continuum of ATP13A2-associated neurological disorders, which are commonly hallmarked by lysosomal and mitochondrial dysfunction. The disease presentation in our patients with hereditary spastic paraplegia was dominated by an adult-onset lower-limb predominant spastic paraparesis. Cognitive impairment was present in most of the cases and ranged from very mild deficits to advanced dementia with fronto-temporal characteristics. Nerve conduction studies revealed involvement of the peripheral motor and sensory nerves. Only one of five patients with hereditary spastic paraplegia showed clinical indication of extrapyramidal involvement in the form of subtle bradykinesia and slight resting tremor. Neuroimaging cranial investigations revealed pronounced vermian and hemispheric cerebellar atrophy. Notably, reduced striatal dopamine was apparent in the brain of one of the patients, who had no clinical signs or symptoms of extrapyramidal involvement.


Subject(s)
Genetic Predisposition to Disease/genetics , Mutation/genetics , Proton-Translocating ATPases/genetics , Spastic Paraplegia, Hereditary/genetics , Adult , Animals , Cells, Cultured/cytology , Cells, Cultured/ultrastructure , Chlorocebus aethiops , Cognition Disorders/etiology , Cognition Disorders/genetics , Enzyme Inhibitors/pharmacology , Family Health , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Leupeptins/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/ultrastructure , Male , Mental Disorders/etiology , Mental Disorders/genetics , Middle Aged , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Neuropsychological Tests , Psychiatric Status Rating Scales , Spastic Paraplegia, Hereditary/complications , Spastic Paraplegia, Hereditary/diagnostic imaging
9.
Am J Hum Genet ; 92(6): 955-64, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23664119

ABSTRACT

The most common form of spinal muscular atrophy (SMA) is a recessive disorder caused by deleterious SMN1 mutations in 5q13, whereas the genetic etiologies of non-5q SMA are very heterogeneous and largely remain to be elucidated. In a Bulgarian family affected by autosomal-dominant proximal SMA, we performed genome-wide linkage analysis and whole-exome sequencing and found a heterozygous de novo c.320C>T (p.Ser107Leu) mutation in bicaudal D homolog 2 (Drosophila) (BICD2). Further analysis of BICD2 in a cohort of 119 individuals with non-5q SMA identified a second de novo BICD2 mutation, c.2321A>G (p.Glu774Gly), in a simplex case. Detailed clinical and electrophysiological investigations revealed that both families are affected by a very similar disease course, characterized by early childhood onset, predominant involvement of lower extremities, and very slow disease progression. The amino acid substitutions are located in two interaction domains of BICD2, an adaptor protein linking the dynein molecular motor with its cargo. Our immunoprecipitation and localization experiments in HeLa and SH-SY5Y cells and affected individuals' lymphoblasts demonstrated that p.Ser107Leu causes increased dynein binding and thus leads to accumulation of BICD2 at the microtubule-organizing complex and Golgi fragmentation. In addition, the altered protein had a reduced colocalization with RAB6A, a regulator of vesicle trafficking between the Golgi and the endoplasmic reticulum. The interaction between p.Glu744Gly altered BICD2 and RAB6A was impaired, which also led to their reduced colocalization. Our study identifies BICD2 mutations as a cause of non-5q linked SMA and highlights the importance of dynein-mediated motility in motor neuron function in humans.


Subject(s)
Carrier Proteins/genetics , Genes, Dominant , Muscular Atrophy, Spinal/genetics , Mutation, Missense , Adult , Base Sequence , Carrier Proteins/metabolism , Child , Child, Preschool , Female , Genetic Association Studies , HeLa Cells , Humans , Male , Microtubule-Associated Proteins , Middle Aged , Muscular Atrophy, Spinal/metabolism , Pedigree , Protein Transport , Sequence Analysis, DNA , Young Adult , rab GTP-Binding Proteins/metabolism
10.
Acta Neuropathol ; 131(4): 505-23, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26744348

ABSTRACT

There is a growing appreciation that membrane-bound organelles in eukaryotic cells communicate directly with one another through direct membrane contact sites. Mitochondria-associated membranes are specialized subdomains of the endoplasmic reticulum that function as membrane contact sites between the endoplasmic reticulum and mitochondria. These sites have emerged as major players in lipid metabolism and calcium signaling. More recently also autophagy and mitochondrial dynamics have been found to be regulated at ER-mitochondria contact sites. Neurons critically depend on mitochondria-associated membranes as a means to exchange metabolites and signaling molecules between these organelles. This is underscored by the fact that genes affecting mitochondrial and endoplasmic reticulum homeostasis are clearly overrepresented in several hereditary neurodegenerative disorders. Conversely, the processes affected by the contact sites between the endoplasmic reticulum and mitochondria are widely implicated in neurodegeneration. This review will focus on the most recent data addressing the structural composition and function of the mitochondria-associated membranes. In addition, the 3D morphology of the contact sites as observed using volume electron microscopy is discussed. Finally, it will highlight the role of several key proteins associated with these contact sites that are involved not only in dementias, amyotrophic lateral sclerosis and Parkinson's disease, but also in axonopathies such as hereditary spastic paraplegia and Charcot-Marie-Tooth disease.


Subject(s)
Endoplasmic Reticulum/metabolism , Mitochondrial Membranes/metabolism , Nerve Degeneration/pathology , Neurons/ultrastructure , Animals , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Humans , Nerve Degeneration/metabolism
11.
Brain ; 138(Pt 11): 3238-50, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26384929

ABSTRACT

The epileptic encephalopathies are a clinically and aetiologically heterogeneous subgroup of epilepsy syndromes. Most epileptic encephalopathies have a genetic cause and patients are often found to carry a heterozygous de novo mutation in one of the genes associated with the disease entity. Occasionally recessive mutations are identified: a recent publication described a distinct neonatal epileptic encephalopathy (MIM 615905) caused by autosomal recessive mutations in the SLC13A5 gene. Here, we report eight additional patients belonging to four different families with autosomal recessive mutations in SLC13A5. SLC13A5 encodes a high affinity sodium-dependent citrate transporter, which is expressed in the brain. Neurons are considered incapable of de novo synthesis of tricarboxylic acid cycle intermediates; therefore they rely on the uptake of intermediates, such as citrate, to maintain their energy status and neurotransmitter production. The effect of all seven identified mutations (two premature stops and five amino acid substitutions) was studied in vitro, using immunocytochemistry, selective western blot and mass spectrometry. We hereby demonstrate that cells expressing mutant sodium-dependent citrate transporter have a complete loss of citrate uptake due to various cellular loss-of-function mechanisms. In addition, we provide independent proof of the involvement of autosomal recessive SLC13A5 mutations in the development of neonatal epileptic encephalopathies, and highlight teeth hypoplasia as a possible indicator for SLC13A5 screening. All three patients who tried the ketogenic diet responded well to this treatment, and future studies will allow us to ascertain whether this is a recurrent feature in this severe disorder.


Subject(s)
Anodontia/genetics , Citric Acid/metabolism , Developmental Disabilities/genetics , Epilepsy/genetics , Symporters/genetics , Adolescent , Brain Diseases/genetics , Child , Female , Genes, Recessive , Genetic Predisposition to Disease , HEK293 Cells , Humans , Male , Mutation , Pedigree , Symporters/metabolism
12.
Neurobiol Dis ; 65: 211-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24521780

ABSTRACT

Charcot-Marie-Tooth disease type 2B (CMT2B) is an inherited axonal peripheral neuropathy. It is characterised by prominent sensory loss, often complicated by severe ulcero-mutilations of toes or feet, and variable motor involvement. Missense mutations in RAB7A, the gene encoding the small GTPase Rab7, cause CMT2B and increase Rab7 activity. Rab7 is ubiquitously expressed and is involved in degradation through the lysosomal pathway. In the neurons, Rab7 plays a role in the long-range retrograde transport of signalling endosomes in the axons. Here we developed the first animal model of CMT2B, modelling one of the mutations (L129F) in Drosophila melanogaster. Behavioural assays show that this model recapitulates several hallmarks of the human disease. Upon expression of mutant Rab7 in the sensory neurons, larvae present with a reduction of temperature and pain perception. Furthermore, the larvae exhibit a crawling defect when the mutant protein is expressed in the motor neurons. Analysis of axonal transport of Rab7 positive vesicles in sensory neurons of Drosophila larvae and in neurites of mammalian neuroblastoma cells demonstrates that mutant vesicles pause less than their wild-type counterparts. This latter finding indicates that alterations in vesicle transport might contribute to the pathomechanism of CMT2B.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/physiopathology , Mutation/genetics , rab GTP-Binding Proteins/genetics , Animals , Animals, Genetically Modified , Behavior, Animal/physiology , Cell Differentiation , Cell Line, Transformed , Dendrites/pathology , Disease Models, Animal , Drosophila , Drosophila Proteins/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Laminopathies , Larva , Male , Motor Activity/genetics , Neuroblastoma/pathology , Sensory Receptor Cells/pathology , Transduction, Genetic , rab7 GTP-Binding Proteins
13.
Neurobiol Dis ; 68: 180-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24807208

ABSTRACT

Aminoacyl-tRNA synthetases are ubiquitously expressed proteins that charge tRNAs with their cognate amino acids. By ensuring the fidelity of protein synthesis, these enzymes are essential for the viability of every cell. Yet, mutations in six tRNA synthetases specifically affect the peripheral nerves and cause Charcot-Marie-Tooth (CMT) disease. The CMT-causing mutations in tyrosyl- and glycyl-tRNA synthetases (YARS and GARS, respectively) alter the activity of the proteins in a range of ways (some mutations do not impact charging function, while others abrogate it), making a loss of function in tRNA charging unlikely to be the cause of disease pathology. It is currently unknown which cellular mechanisms are triggered by the mutant enzymes and how this leads to neurodegeneration. Here, by expressing two pathogenic mutations (G240R, P234KY) in Drosophila, we generated a model for GARS-associated neuropathy. We observed compromised viability, and behavioral, electrophysiological and morphological impairment in flies expressing the cytoplasmic isoform of mutant GARS. Their features recapitulated several hallmarks of CMT pathophysiology and were similar to the phenotypes identified in our previously described Drosophila model of YARS-associated neuropathy. Furthermore, CG8316 and CG15599 - genes identified in a retinal degeneration screen to modify mutant YARS, also modified the mutant GARS phenotypes. Our study presents genetic evidence for common mutant-specific interactions between two CMT-associated aminoacyl-tRNA synthetases, lending support for a shared mechanism responsible for the synthetase-induced peripheral neuropathies.


Subject(s)
Charcot-Marie-Tooth Disease/complications , Charcot-Marie-Tooth Disease/genetics , Glycine-tRNA Ligase/genetics , Mutation/genetics , Peripheral Nervous System Diseases/etiology , Tyrosine-tRNA Ligase/genetics , Animals , Animals, Genetically Modified , Charcot-Marie-Tooth Disease/pathology , Dextrans , Disease Models, Animal , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Humans , Male , Membrane Potentials/genetics , Membrane Potentials/physiology , Nerve Fibers/physiology , Neurons/pathology , Neurons/physiology , Peripheral Nervous System Diseases/genetics , Retina/pathology , Retina/ultrastructure , Retinal Degeneration/diagnosis , Retinal Degeneration/etiology , Retinal Degeneration/genetics , Rhodamines , Wings, Animal/pathology , Wings, Animal/ultrastructure
14.
Mol Neurodegener ; 19(1): 37, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38654375

ABSTRACT

BACKGROUND: Microglia play important roles in maintaining brain homeostasis and neurodegeneration. The discovery of genetic variants in genes predominately or exclusively expressed in myeloid cells, such as Apolipoprotein E (APOE) and triggering receptor expressed on myeloid cells 2 (TREM2), as the strongest risk factors for Alzheimer's disease (AD) highlights the importance of microglial biology in the brain. The sequence, structure and function of several microglial proteins are poorly conserved across species, which has hampered the development of strategies aiming to modulate the expression of specific microglial genes. One way to target APOE and TREM2 is to modulate their expression using antisense oligonucleotides (ASOs). METHODS: In this study, we identified, produced, and tested novel, selective and potent ASOs for human APOE and TREM2. We used a combination of in vitro iPSC-microglia models, as well as microglial xenotransplanted mice to provide proof of activity in human microglial in vivo. RESULTS: We proved their efficacy in human iPSC microglia in vitro, as well as their pharmacological activity in vivo in a xenografted microglia model. We demonstrate ASOs targeting human microglia can modify their transcriptional profile and their response to amyloid-ß plaques in vivo in a model of AD. CONCLUSIONS: This study is the first proof-of-concept that human microglial can be modulated using ASOs in a dose-dependent manner to manipulate microglia phenotypes and response to neurodegeneration in vivo.


Subject(s)
Alzheimer Disease , Microglia , Oligonucleotides, Antisense , Microglia/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Humans , Oligonucleotides, Antisense/pharmacology , Animals , Mice , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Induced Pluripotent Stem Cells/metabolism , Gene Expression Regulation/drug effects , Disease Models, Animal
15.
Magn Reson Med ; 69(4): 1115-21, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23494926

ABSTRACT

PURPOSE: Amyloid deposition in the brain is considered an initial event in the progression of Alzheimer's disease. We hypothesized that the presence of amyloid plaques in the brain of APP/presenilin 1 mice leads to higher diffusion kurtosis measures due to increased microstructural complexity. As such, our purpose was to provide an in vivo proof of principle for detection of amyloidosis by diffusion kurtosis imaging (DKI). METHODS: APPKM670/671NL /presenilin 1 L166P mice (n = 5) and wild-type littermates (n = 5) underwent DKI at the age of 16 months. Averaged diffusion and diffusion kurtosis parameters were obtained for multiple regions (hippocampus-cortex-thalamus-cerebellum). After DKI, mice were sacrificed for amyloid staining. RESULTS: Histograms of the frequency distribution of the DKI parameters tended to shift to higher values. After normalization of absolute values to the cerebellum, a nearly plaque-free region, mean, radial, and axial diffusion kurtosis were significantly higher in APP/presenilin 1 mice as compared to wild-type in the cortex and thalamus, regions demonstrating substantial amyloid staining. CONCLUSION: The current study, although small-scale, suggests increased DKI metrics, in the absence of alterations in diffusion tensor imaging metrics in the cortex and thalamus of APP/presenilin 1 mice with established amyloidosis. These results warrant further investigations on the potential of DKI as a sensitive marker for Alzheimer's disease.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloidosis/pathology , Diffusion Magnetic Resonance Imaging/methods , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Presenilin-1/genetics , Algorithms , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloidosis/complications , Amyloidosis/genetics , Animals , Mice , Mice, Transgenic , Reproducibility of Results , Sensitivity and Specificity
16.
Acta Neuropathol ; 126(1): 93-108, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23728742

ABSTRACT

Mutations in the small heat shock protein HSPB1 (HSP27) are a cause of axonal Charcot-Marie-Tooth neuropathy (CMT2F) and distal hereditary motor neuropathy. To better understand the effect of mutations in HSPB1 on the neuronal cytoskeleton, we stably transduced neuronal cells with wild-type and mutant HSPB1 and investigated axonal transport of neurofilaments (NFs). We observed that mutant HSPB1 affected the binding of NFs to the anterograde motor protein kinesin, reducing anterograde transport of NFs. These deficits were associated with an increased phosphorylation of NFs and cyclin-dependent kinase Cdk5. As Cdk5 mediates NF phosphorylation, inhibition of Cdk5/p35 restored NF phosphorylation level, as well as NF binding to kinesin in mutant HSPB1 neuronal cells. Altogether, we demonstrate that HSPB1 mutations induce hyperphosphorylation of NFs through Cdk5 and reduce anterograde transport of NFs.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Cyclin-Dependent Kinase 5/metabolism , HSP27 Heat-Shock Proteins/genetics , Mutation/genetics , Neurofilament Proteins/metabolism , Axonal Transport/genetics , Axons/metabolism , Axons/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase 5/genetics , Heat-Shock Proteins , Humans , Immunoprecipitation , Kinesins/metabolism , Molecular Chaperones , Neuroblastoma/pathology , Phosphorylation/genetics , Transfection/methods
17.
Nat Cell Biol ; 25(3): 467-480, 2023 03.
Article in English | MEDLINE | ID: mdl-36690850

ABSTRACT

Mitochondria are complex organelles with different compartments, each harbouring their own protein quality control factors. While chaperones of the mitochondrial matrix are well characterized, it is poorly understood which chaperones protect the mitochondrial intermembrane space. Here we show that cytosolic small heat shock proteins are imported under basal conditions into the mitochondrial intermembrane space, where they operate as molecular chaperones. Protein misfolding in the mitochondrial intermembrane space leads to increased recruitment of small heat shock proteins. Depletion of small heat shock proteins leads to mitochondrial swelling and reduced respiration, while aggregation of aggregation-prone substrates is countered in their presence. Charcot-Marie-Tooth disease-causing mutations disturb the mitochondrial function of HSPB1, potentially linking previously observed mitochondrial dysfunction in Charcot-Marie-Tooth type 2F to its role in the mitochondrial intermembrane space. Our results reveal that small heat shock proteins form a chaperone system that operates in the mitochondrial intermembrane space.


Subject(s)
Heat-Shock Proteins, Small , Heat-Shock Proteins, Small/genetics , Heat-Shock Proteins, Small/metabolism , Molecular Chaperones/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
18.
Autophagy ; 19(8): 2217-2239, 2023 08.
Article in English | MEDLINE | ID: mdl-36854646

ABSTRACT

Chaperone-assisted selective autophagy (CASA) is a highly selective pathway for the disposal of misfolding and aggregating proteins. In muscle, CASA assures muscle integrity by favoring the turnover of structural components damaged by mechanical strain. In neurons, CASA promotes the removal of aggregating substrates. A crucial player of CASA is HSPB8 (heat shock protein family B (small) member 8), which acts in a complex with HSPA, their cochaperone BAG3, and the E3 ubiquitin ligase STUB1. Recently, four novel HSPB8 frameshift (fs) gene mutations have been linked to neuromyopathies, and encode carboxy-terminally mutated HSPB8, sharing a common C-terminal extension. Here, we analyzed the biochemical and functional alterations associated with the HSPB8_fs mutant proteins. We demonstrated that HSPB8_fs mutants are highly insoluble and tend to form proteinaceous aggregates in the cytoplasm. Notably, all HSPB8 frameshift mutants retain their ability to interact with CASA members but sequester them into the HSPB8-positive aggregates together with two autophagy receptors SQSTM1/p62 and TAX1BP1. This copartitioning process negatively affects the CASA capability to remove its clients and causes a general failure in proteostasis response. Further analyses revealed that the aggregation of the HSPB8_fs mutants occurs independently of the other CASA members or from the autophagy receptors interaction, but it is an intrinsic feature of the mutated amino acid sequence. HSPB8_fs mutants aggregation alters the differentiation capacity of muscle cells and impairs sarcomere organization. Collectively, these results shed light on a potential pathogenic mechanism shared by the HSPB8_fs mutants described in neuromuscular diseases.Abbreviations : ACD: α-crystallin domain; ACTN: actinin alpha; BAG3: BAG cochaperone 3; C: carboxy; CASA: chaperone-assisted selective autophagy; CE: carboxy-terminal extension; CLEM: correlative light and electron microscopy; CMT2L: Charcot-Marie-Tooth type 2L; CTR: carboxy-terminal region; dHMNII: distal hereditary motor neuropathy type II; EV: empty vector; FRA: filter retardation assay; fs: frameshift; HSPA/HSP70: heat shock protein family A (Hsp70); HSPB1/Hsp27: heat shock protein family B (small) member 1; HSPB8/Hsp22: heat shock protein family B (small) member 8; HTT: huntingtin; KO: knockout; MAP1LC3B/LC3: microtubule associated protein 1 light chain 3 beta; MD: molecular dynamics; MTOC: microtubule organizing center; MYH: myosin heavy chain; MYOG: myogenin; NBR1: NBR1 autophagy cargo receptor; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; NSC34: Neuroblastoma X Spinal Cord 34; OPTN: optineurin; polyQ: polyglutamine; SQSTM1/p62: sequestosome 1; STUB1/CHIP: STIP1 homology and U-box containing protein 1; TARDBP/TDP-43: TAR DNA binding protein; TAX1BP1: Tax1 binding protein 1; TUBA: tubulin alpha; WT: wild-type.


Subject(s)
Charcot-Marie-Tooth Disease , Neuromuscular Diseases , Humans , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Autophagy/genetics , Heat-Shock Proteins/metabolism , Charcot-Marie-Tooth Disease/genetics , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism
19.
Nat Commun ; 14(1): 999, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36890170

ABSTRACT

Dominant mutations in tyrosyl-tRNA synthetase (YARS1) and six other tRNA ligases cause Charcot-Marie-Tooth peripheral neuropathy (CMT). Loss of aminoacylation is not required for their pathogenicity, suggesting a gain-of-function disease mechanism. By an unbiased genetic screen in Drosophila, we link YARS1 dysfunction to actin cytoskeleton organization. Biochemical studies uncover yet unknown actin-bundling property of YARS1 to be enhanced by a CMT mutation, leading to actin disorganization in the Drosophila nervous system, human SH-SY5Y neuroblastoma cells, and patient-derived fibroblasts. Genetic modulation of F-actin organization improves hallmark electrophysiological and morphological features in neurons of flies expressing CMT-causing YARS1 mutations. Similar beneficial effects are observed in flies expressing a neuropathy-causing glycyl-tRNA synthetase. Hence, in this work, we show that YARS1 is an evolutionary-conserved F-actin organizer which links the actin cytoskeleton to tRNA-synthetase-induced neurodegeneration.


Subject(s)
Actins , Tyrosine-tRNA Ligase , Animals , Humans , Actins/metabolism , Charcot-Marie-Tooth Disease/genetics , Drosophila/genetics , Glycine-tRNA Ligase/genetics , Mutation , RNA, Transfer , Tyrosine-tRNA Ligase/genetics , Tyrosine-tRNA Ligase/metabolism , Cell Line, Tumor
20.
J Neurosci ; 31(43): 15320-8, 2011 Oct 26.
Article in English | MEDLINE | ID: mdl-22031878

ABSTRACT

Mutations in the small heat shock protein HSPB1 (HSP27) are causative for Charcot-Marie-Tooth (CMT) neuropathy. We previously showed that a subset of these mutations displays higher chaperone activity and enhanced affinity to client proteins. We hypothesized that this excessive binding property might cause the HSPB1 mutant proteins to disturb the function of proteins essential for the maintenance or survival of peripheral neurons. In the present work, we explored this hypothesis further and compared the protein complexes formed by wild-type and mutant HSPB1. Tubulin came out as the most striking differential interacting protein, with hyperactive mutants binding more strongly to both tubulin and microtubules. This anomalous binding leads to a stabilization of the microtubule network in a microtubule-associated protein-like manner as reflected by resistance to cold depolymerization, faster network recovery after nocodazole treatment, and decreased rescue and catastrophe rates of individual microtubules. In a transgenic mouse model for mutant HSPB1 that recapitulates all features of CMT, we could confirm the enhanced interaction of mutant HSPB1 with tubulin. Increased stability of the microtubule network was also clear in neurons isolated from these mice. Since neuronal cells are particularly vulnerable to disturbances in microtubule dynamics, this mechanism might explain the neuron-specific CMT phenotype caused by HSPB1 mutations.


Subject(s)
HSP27 Heat-Shock Proteins/genetics , Microtubules/metabolism , Mutation/genetics , Neurons/metabolism , Analysis of Variance , Animals , Cells, Cultured , Chlorocebus aethiops , Ganglia, Spinal/cytology , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Heat-Shock Proteins , Humans , Ice/adverse effects , Mice , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Molecular Chaperones , Neurons/drug effects , Nocodazole/pharmacology , Protein Binding , Surface Plasmon Resonance , Tandem Mass Spectrometry/methods , Time Factors , Transfection/methods , Tubulin/genetics , Tubulin/pharmacology , Tubulin Modulators/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL