Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mol Genet ; 33(12): 1055-1063, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38493358

ABSTRACT

Claudin-25 (CLDN-25), also known as Claudin containing domain 1, is an uncharacterized claudin family member. It has less conserved amino acid sequences when compared to other claudins. It also has a very broad tissue expression profile and there is currently a lack of functional information from murine knockout models. Here, we report a de novo missense heterozygous variant in CLDN25 (c. 745G>C, p. A249P) found in a patient diagnosed with Pelizaeus-Merzbacher-like leukodystrophy and presenting with symptoms such as delayed motor development, several episodes of tonic absent seizures and generalized dystonia. The variant protein does not localize to the cell-cell borders where it would normally be expected to be expressed. Amino acid position 249 is located 4 amino acids from the C-terminal end of the protein where most claudin family members have a conserved binding motif for the key scaffolding protein ZO-1. However, CLDN-25 does not contain this motif. Here, we show that the C-terminal end of CLDN-25 is required for its junctional localization in a ZO-1 independent manner. The A249P mutant protein as well as a deletion mutant lacking its last 5 C-terminal amino acids also failed to localize to the cell-cell border in vitro. Intriguingly, cellular knockout of CLDN25, in vitro, appeared to increase the integrity of the tight junction between 2 contacting cells, while driving highly unusual increased movement of solutes between cells. We propose that the barrier function of CLDN-25 is akin to a decoy claudin, whereby decreasing its expression in "leaky" epithelial cells and endothelial cells will drive dynamic changes in the adhesion and interaction capacity of cell-cell contact points. While it remains unclear how this de novo CLDN-25 mutant induces leukodystrophy, our findings strongly suggest that this mutation induces haploinsufficiency of CLDN-25. Elucidating the function of this uncharacterized claudin protein will lead to a better understanding of the role of claudin proteins in health and disease.


Subject(s)
Claudins , Pelizaeus-Merzbacher Disease , Humans , Claudins/genetics , Claudins/metabolism , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/metabolism , Loss of Function Mutation , Male , Mutation, Missense , Female , Animals , Amino Acid Sequence
2.
Nature ; 585(7825): 397-403, 2020 09.
Article in English | MEDLINE | ID: mdl-32610343

ABSTRACT

Mutations in PLP1, the gene that encodes proteolipid protein (PLP), result in failure of myelination and neurological dysfunction in the X-chromosome-linked leukodystrophy Pelizaeus-Merzbacher disease (PMD)1,2. Most PLP1 mutations, including point mutations and supernumerary copy variants, lead to severe and fatal disease. Patients who lack PLP1 expression, and Plp1-null mice, can display comparatively mild phenotypes, suggesting that PLP1 suppression might provide a general therapeutic strategy for PMD1,3-5. Here we show, using CRISPR-Cas9 to suppress Plp1 expression in the jimpy (Plp1jp) point-mutation mouse model of severe PMD, increased myelination and restored nerve conduction velocity, motor function and lifespan of the mice to wild-type levels. To evaluate the translational potential of this strategy, we identified antisense oligonucleotides that stably decrease the levels of Plp1 mRNA and PLP protein throughout the neuraxis in vivo. Administration of a single dose of Plp1-targeting antisense oligonucleotides in postnatal jimpy mice fully restored oligodendrocyte numbers, increased myelination, improved motor performance, normalized respiratory function and extended lifespan up to an eight-month end point. These results suggest that PLP1 suppression could be developed as a treatment for PMD in humans. More broadly, we demonstrate that oligonucleotide-based therapeutic agents can be delivered to oligodendrocytes in vivo to modulate neurological function and lifespan, establishing a new pharmaceutical modality for myelin disorders.


Subject(s)
Disease Models, Animal , Myelin Proteolipid Protein/deficiency , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/therapy , Animals , CRISPR-Cas Systems , Female , Gene Editing , Hypoxia/metabolism , Male , Mice , Mice, Mutant Strains , Motor Activity/genetics , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Pelizaeus-Merzbacher Disease/metabolism , Point Mutation , Respiratory Function Tests , Survival Analysis
3.
Development ; 149(20)2022 10 15.
Article in English | MEDLINE | ID: mdl-35748297

ABSTRACT

Oligodendrocytes, the myelinating cells of the central nervous system, possess great potential for disease modeling and cell transplantation-based therapies for leukodystrophies. However, caveats to oligodendrocyte differentiation protocols ( Ehrlich et al., 2017; Wang et al., 2013; Douvaras and Fossati, 2015) from human embryonic stem and induced pluripotent stem cells (iPSCs), which include slow and inefficient differentiation, and tumorigenic potential of contaminating undifferentiated pluripotent cells, are major bottlenecks towards their translational utility. Here, we report the rapid generation of human oligodendrocytes by direct lineage conversion of human dermal fibroblasts (HDFs). We show that the combination of the four transcription factors OLIG2, SOX10, ASCL1 and NKX2.2 is sufficient to convert HDFs to induced oligodendrocyte precursor cells (iOPCs). iOPCs resemble human primary and iPSC-derived OPCs based on morphology and transcriptomic analysis. Importantly, iOPCs can differentiate into mature myelinating oligodendrocytes in vitro and in vivo. Finally, iOPCs derived from patients with Pelizaeus Merzbacher disease, a hypomyelinating leukodystrophy caused by mutations in the proteolipid protein 1 (PLP1) gene, showed increased cell death compared with iOPCs from healthy donors. Thus, human iOPCs generated by direct lineage conversion represent an attractive new source for human cell-based disease models and potentially myelinating cell grafts.


Subject(s)
Induced Pluripotent Stem Cells , Pelizaeus-Merzbacher Disease , Cell Differentiation/physiology , Fibroblasts , Humans , Induced Pluripotent Stem Cells/metabolism , Oligodendroglia/metabolism , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/metabolism , Pelizaeus-Merzbacher Disease/therapy
4.
Mol Cell Neurosci ; 120: 103716, 2022 05.
Article in English | MEDLINE | ID: mdl-35276347

ABSTRACT

Pelizaeus-Merzbacher-like disease type 1 (PMLD1) is a hypomyelinating disorder arising in patients with mutations in GJC2, encoding Connexin47 (Cx47). PMLD1 causes nystagmus, cerebellar ataxia, spasticity and changes in CNS white matter detected by MRI. At least one mutation (p.I33M) yields a much milder phenotype, spastic paraplegia type 44 (SPG44). Cx47 contributes to gap junction communication channels between oligodendrocytes (OLs), the myelinating cells in the central nervous system (CNS), and between OLs and astrocytes. Prior studies in cell lines have shown that PMLD1 mutants such as p.P87S display defective protein trafficking, intracellular retention in the ER and loss-of-function. Here we show that when expressed in primary OLs, three PMLD1 associated mutants (p.P87S, p.Y269D and p.M283T) show ER retention of Cx47 and evidence of activation of the cellular stress (unfolded protein response, UPR) and apoptotic pathways. On the other hand, the milder SPG44 associated mutation p.I33M shows a wild-type-like subcellular distribution and no activation of the UPR or apoptotic pathways. These studies provide new insight into a potential element of toxic gain of function underlying the mechanism of PMLD1 that should help guide future therapeutic approaches.


Subject(s)
Demyelinating Diseases , Lysosomal Storage Diseases , Neurodegenerative Diseases , Pelizaeus-Merzbacher Disease , Connexins/genetics , Connexins/metabolism , Demyelinating Diseases/metabolism , Gap Junctions/genetics , Gap Junctions/metabolism , Humans , Lysosomal Storage Diseases/metabolism , Mutation , Neurodegenerative Diseases/metabolism , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/metabolism , Unfolded Protein Response/genetics
5.
J Hum Genet ; 66(10): 1035-1037, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33785861

ABSTRACT

Monoallelic mutations on TMEM63A have been recently reported as cause of a previously unrecognized disorder named "infantile-onset transient hypomyelination". Clinical and neuroradiological presentation is described as highly similar to Pelizaeus-Merzbacher Disease but evolution over time was surprisingly benign with a progressive spontaneous improving course. We report on a new TMEM63A-mutated girl. The clinical picture was similar to the one already described except for the presence of recurrent episodes of unilateral eyelid twitching, and for the evidence of spinal cord involvement on MRI. These are interesting findings helping in distinguishing this condition from classic PMD since early disease stages. However, additional observations are needed to confirm if these are common features of this condition.


Subject(s)
Genetic Predisposition to Disease , Membrane Proteins/genetics , Myelin Proteolipid Protein/genetics , Pelizaeus-Merzbacher Disease/genetics , Spinal Cord/diagnostic imaging , Female , Humans , Infant , Magnetic Resonance Imaging , Mutation/genetics , Pelizaeus-Merzbacher Disease/diagnostic imaging , Pelizaeus-Merzbacher Disease/metabolism , Pelizaeus-Merzbacher Disease/pathology , Spinal Cord/metabolism , Spinal Cord/pathology
6.
Int J Mol Sci ; 22(11)2021 May 30.
Article in English | MEDLINE | ID: mdl-34070744

ABSTRACT

The ClC-2 channel plays a critical role in maintaining ion homeostasis in the brain and the testis. Loss-of-function mutations in the ClC-2-encoding human CLCN2 gene are linked to the white matter disease leukodystrophy. Clcn2-deficient mice display neuronal myelin vacuolation and testicular degeneration. Leukodystrophy-causing ClC-2 mutant channels are associated with anomalous proteostasis manifesting enhanced endoplasmic reticulum (ER)-associated degradation. The molecular nature of the ER quality control system for ClC-2 protein remains elusive. In mouse testicular tissues and Leydig cells, we demonstrated that endogenous ClC-2 co-existed in the same protein complex with the molecular chaperones heat shock protein 90ß (Hsp90ß) and heat shock cognate protein (Hsc70), as well as the associated co-chaperones Hsp70/Hsp90 organizing protein (HOP), activator of Hsp90 ATPase homolog 1 (Aha1), and FK506-binding protein 8 (FKBP8). Further biochemical analyses revealed that the Hsp90ß-Hsc70 chaperone/co-chaperone system promoted mouse and human ClC-2 protein biogenesis. FKBP8 additionally facilitated membrane trafficking of ClC-2 channels. Interestingly, treatment with the Hsp90-targeting small molecule 17-allylamino-17-demethoxygeldanamycin (17-AAG) substantially boosted ClC-2 protein expression. Also, 17-AAG effectively increased both total and cell surface protein levels of leukodystrophy-causing loss-of-function ClC-2 mutant channels. Our findings highlight the therapeutic potential of 17-AAG in correcting anomalous ClC-2 proteostasis associated with leukodystrophy.


Subject(s)
Brain/metabolism , Chloride Channels/genetics , Leydig Cells/metabolism , Neurons/metabolism , Pelizaeus-Merzbacher Disease/genetics , Proteostasis/genetics , Animals , Benzoquinones/pharmacology , Brain/drug effects , Brain/pathology , CHO Cells , CLC-2 Chloride Channels , Chloride Channels/deficiency , Cricetulus , Disease Models, Animal , Endoplasmic Reticulum-Associated Degradation/drug effects , Gene Expression Regulation , HEK293 Cells , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic/pharmacology , Leydig Cells/drug effects , Leydig Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Neurons/drug effects , Neurons/pathology , Pelizaeus-Merzbacher Disease/drug therapy , Pelizaeus-Merzbacher Disease/metabolism , Pelizaeus-Merzbacher Disease/pathology , Protein Isoforms/deficiency , Protein Isoforms/genetics , Signal Transduction , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
7.
Acta Neurol Scand ; 139(2): 135-142, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30192380

ABSTRACT

OBJECTIVE: LMNB1-related autosomal dominant leukodystrophy is caused by an overexpression of the protein lamin B1, usually due to a duplication of the LMNB1 gene. Symptoms start in 5th to 6th decade. This slowly progressive disease terminates with death. We studied brain glucose metabolism in this disease using 18 F-fluorodeoxyglucose positron emission tomography (PET). METHODS: We examined 8 patients, aged 48-64 years, in varying stages of clinical symptomatology. Two patients were investigated with quantitative PET on clinical indications after which six more patients were recruited. Absolute glucose metabolism was analyzed with the PVElab software in 6 patients and 18 healthy controls. A semiquantitative analysis using the CortexID software was performed in seven investigations, relating local metabolism levels to global glucose metabolism. RESULTS: The clinical quantitative PET revealed low global glucose metabolism, with the most marked reduction in the cerebellum. In the PVElab analysis, patients presented low mean glucose metabolism in the cerebellum, brainstem and global grey matter. In the semiquantitative analysis, 2 patients showed a decreased metabolism in the cerebellum and 4 patients a relatively higher metabolism in parts of the temporal lobes. Since none of the patients showed an increased metabolism in the quantitative analysis, we interpret these increases as "pseudo-increases" related to a globally reduced metabolism. CONCLUSIONS: Global reduction of grey matter glucose metabolism in this white matter disease most likely depends on a combination of cortical afferent dysfunction and, in later stages, neuronal loss. The lowest metabolism in the cerebellum is consistent with histopathological findings and prominent cerebellar symptoms.


Subject(s)
Cerebellum/diagnostic imaging , Lamin Type B/genetics , Pelizaeus-Merzbacher Disease/diagnostic imaging , Positron-Emission Tomography , Cerebellum/metabolism , Cerebellum/pathology , Female , Fluorodeoxyglucose F18 , Humans , Male , Middle Aged , Pelizaeus-Merzbacher Disease/metabolism , Pelizaeus-Merzbacher Disease/pathology , Radiopharmaceuticals
8.
Hum Mol Genet ; 24(10): 2746-56, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25637521

ABSTRACT

Adult-onset autosomal dominant leukodystrophy (ADLD) is a slowly progressive neurological disorder characterized by autonomic dysfunction, followed by cerebellar and pyramidal features. ADLD is caused by duplication of the lamin B1 gene (LMNB1), which leads to its increased expression. The molecular pathways involved in the disease are still poorly understood. Hence, we analyzed global gene expression in fibroblasts and whole blood of LMNB1 duplication carriers and used Gene Set Enrichment Analysis to explore their gene signatures. We found that LMNB1 duplication is associated with dysregulation of genes involved in the immune system, neuronal and skeletal development. Genes with an altered transcriptional profile clustered in specific genomic regions. Among the dysregulated genes, we further studied the role of RAVER2, which we found to be overexpressed at mRNA and protein level. RAVER2 encodes a putative trans regulator of the splicing repressor polypyrimidine tract binding protein (PTB) and is likely implicated in alternative splicing regulation. Functional studies demonstrated an abnormal splicing pattern of several PTB-target genes and of the myelin protein gene PLP1, previously demonstrated to be involved in ADLD. Mutant mice with different lamin B1 expression levels confirmed that Raver2 expression is dependent on lamin B1 in neural tissue and determines an altered splicing pattern of PTB-target genes and Plp1. Overall our results demonstrate that deregulation of lamin B1 expression induces modified splicing of several genes, likely driven by raver-2 overexpression, and suggest that an alteration of mRNA processing could be a pathogenic mechanism in ADLD.


Subject(s)
Alternative Splicing , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Lamin Type B/genetics , Pelizaeus-Merzbacher Disease/genetics , Animals , Fibroblasts/metabolism , Gene Duplication , Gene Expression Profiling , Humans , Leukocytes/metabolism , Mice , Mice, Knockout , Pelizaeus-Merzbacher Disease/metabolism , Up-Regulation
9.
Biochim Biophys Acta ; 1851(8): 1083-94, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25724171

ABSTRACT

The largest pool of free cholesterol in mammals resides in myelin membranes. Myelin facilitates rapid saltatory impulse propagation by electrical insulation of axons. This function is achieved by ensheathing axons with a tightly compacted stack of membranes. Cholesterol influences myelination at many steps, from the differentiation of myelinating glial cells, over the process of myelin membrane biogenesis, to the functionality of mature myelin. Cholesterol emerged as the only integral myelin component that is essential and rate-limiting for the development of myelin in the central and peripheral nervous system. Moreover, disorders that interfere with sterol synthesis or intracellular trafficking of cholesterol and other lipids cause hypomyelination and neurodegeneration. This review summarizes recent results on the roles of cholesterol in CNS myelin biogenesis in normal development and under different pathological conditions. This article is part of a Special Issue entitled Brain Lipids.


Subject(s)
Cholesterol/metabolism , Myelin Sheath/metabolism , Niemann-Pick Disease, Type C/metabolism , Pelizaeus-Merzbacher Disease/metabolism , Smith-Lemli-Opitz Syndrome/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Central Nervous System/cytology , Central Nervous System/metabolism , Homeostasis , Humans , Myelin Sheath/chemistry , Myelin Sheath/ultrastructure , Neurons/cytology , Neurons/metabolism , Niemann-Pick Disease, Type C/pathology , Pelizaeus-Merzbacher Disease/pathology , Peripheral Nervous System/cytology , Peripheral Nervous System/metabolism , Smith-Lemli-Opitz Syndrome/pathology , Synaptic Transmission
10.
Clin Genet ; 90(4): 293-304, 2016 10.
Article in English | MEDLINE | ID: mdl-27234264

ABSTRACT

Hypomyelinating leukodystrophies (HLDs) are a group of neurodevelopmental disorders that affect proper formation of the myelin sheath in the central nervous system. They are characterized by developmental delay, hypotonia, spasticity, and variable intellectual disability. In the past various classification systems for HLDs have been used, based on imaging findings, clinical manifestation, and organelle-specific disorders. Here we present a molecular insight into HLDs based on a defect in specific gene engaged in myelination. We discuss recent findings on pathogenesis, clinical presentation, and imaging related to these disorders. We focus on HLDs that are in use in differential diagnostics of Pelizaeus-Merzbacher disease (PMD), with a special emphasis on Allan-Herndon-Dudley syndrome (AHDS), an X-linked condition with delayed myelination due to thyroid transport disturbances. On the background of previously published patients we describe a proband initially considered as presenting with a severe PMD, whose diagnosis of AHDS due to a novel nonsense SLC16A2 mutation unraveled two previously undiagnosed generations of affected males who died in infancy from unexplained reasons. Since AHDS is found to be a relatively frequent cause of X-linked intellectual disability, we emphasize the need for determining the whole thyroid profile especially in hypotonic males with a delay of psychomotor development.


Subject(s)
Leukoencephalopathies/genetics , Mental Retardation, X-Linked/genetics , Muscle Hypotonia/genetics , Muscular Atrophy/genetics , Pelizaeus-Merzbacher Disease/genetics , Child , Child, Preschool , Diagnosis, Differential , Genes, X-Linked , Humans , Leukoencephalopathies/diagnosis , Leukoencephalopathies/metabolism , Male , Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/metabolism , Muscle Hypotonia/diagnosis , Muscle Hypotonia/metabolism , Muscular Atrophy/diagnosis , Muscular Atrophy/metabolism , Mutation , Myelin Sheath/genetics , Myelin Sheath/metabolism , Pedigree , Pelizaeus-Merzbacher Disease/diagnosis , Pelizaeus-Merzbacher Disease/metabolism
11.
FASEB J ; 28(9): 3906-18, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24858279

ABSTRACT

The architecture and structural mechanics of the cell nucleus are defined by the nuclear lamina, which is formed by A- and B-type lamins. Recently, gene duplication and protein overexpression of lamin B1 (LB1) have been reported in pedigrees with autosomal dominant leukodystrophy (ADLD). However, how the overexpression of LB1 affects nuclear mechanics and function and how it may result in pathology remain unexplored. Here, we report that in primary human skin fibroblasts derived from ADLD patients, LB1, but not other lamins, is overexpressed at the nuclear lamina and specifically enhances nuclear stiffness. Transient transfection of LB1 in HEK293 and neuronal N2a cells mimics the mechanical phenotype of ADLD nuclei. Notably, in ADLD fibroblasts, reducing LB1 protein levels by shRNA knockdown restores elasticity values to those indistinguishable from control fibroblasts. Moreover, isolated nuclei from ADLD fibroblasts display a reduced nuclear ion channel open probability on voltage-step application, suggesting that biophysical changes induced by LB1 overexpression may alter nuclear signaling cascades in somatic cells. Overall, the overexpression of LB1 in ADLD cells alters nuclear mechanics and is linked to changes in nuclear signaling, which could help explain the pathogenesis of this disease.


Subject(s)
Cell Nucleus/pathology , Embryo, Mammalian/cytology , Fibroblasts/pathology , Lamin Type B/metabolism , Pelizaeus-Merzbacher Disease/pathology , Skin/cytology , Adult , Animals , Blotting, Western , Case-Control Studies , Cell Membrane Permeability , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Embryo, Mammalian/metabolism , Female , Fibroblasts/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Lamin Type B/antagonists & inhibitors , Lamin Type B/genetics , Male , Mice , Middle Aged , Patch-Clamp Techniques , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/metabolism , Phenotype , RNA, Small Interfering/genetics , Skin/metabolism
12.
J Biol Chem ; 288(11): 7451-7466, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23344956

ABSTRACT

Missense mutations in the proteolipid protein 1 (PLP1) gene cause a wide spectrum of hypomyelinating disorders, from mild spastic paraplegia type 2 to severe Pelizaeus-Merzbacher disease (PMD). Mutant PLP1 accumulates in the endoplasmic reticulum (ER) and induces ER stress. However, the link between the clinical severity of PMD and the cellular response induced by mutant PLP1 remains largely unknown. Accumulation of misfolded proteins in the ER generally leads to up-regulation of ER chaperones to alleviate ER stress. Here, we found that expression of the PLP1-A243V mutant, which causes severe disease, depletes some ER chaperones with a KDEL (Lys-Asp-Glu-Leu) motif, in HeLa cells, MO3.13 oligodendrocytic cells, and primary oligodendrocytes. The same PLP1 mutant also induces fragmentation of the Golgi apparatus (GA). These organelle changes are less prominent in cells with milder disease-associated PLP1 mutants. Similar changes are also observed in cells expressing another disease-causing gene that triggers ER stress, as well as in cells treated with brefeldin A, which induces ER stress and GA fragmentation by inhibiting GA to ER trafficking. We also found that mutant PLP1 disturbs localization of the KDEL receptor, which transports the chaperones with the KDEL motif from the GA to the ER. These data show that PLP1 mutants inhibit GA to ER trafficking, which reduces the supply of ER chaperones and induces GA fragmentation. We propose that depletion of ER chaperones and GA fragmentation induced by mutant misfolded proteins contribute to the pathogenesis of inherited ER stress-related diseases and affect the disease severity.


Subject(s)
Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Golgi Apparatus/metabolism , Molecular Chaperones/metabolism , Mutation , Myelin Proteolipid Protein/metabolism , Pelizaeus-Merzbacher Disease/metabolism , Amino Acid Motifs , Animals , Biotinylation , Disease Models, Animal , HeLa Cells , Humans , Mice , Mutation, Missense , Neuroglia/cytology , Oligodendroglia/cytology , Organelles/metabolism , Protein Transport , Protein Unfolding
13.
Biochim Biophys Acta ; 1832(3): 411-20, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23261988

ABSTRACT

Adult-onset autosomal dominant leukodystrophy (ADLD) is a slowly progressive neurological disorder characterised by pyramidal, cerebellar, and autonomic disturbances. Duplication of the LMNB1 gene is the genetic cause of ADLD, yet the pathogenetic mechanism is not defined. In this study, we analysed cells and muscle tissue from three patients affected by ADLD, carrying an extra copy of the LMNB1 gene. Lamin B1 levels were dramatically increased in ADLD nuclei, both in skin fibroblasts and skeletal muscle fibres. Since lamin B1 is known to bind Oct-1, a transcription factor involved in the oxidative stress pathway, we investigated Oct-1 fate in ADLD. Oct-1 recruitment to the nuclear periphery was increased in ADLD cells, while nucleoplasmic localisation of the transcription factor under oxidative stress conditions was reduced. Importantly, lamin B1 degradation occurring in some, but not all ADLD cell lines, slowed down lamin B1 and Oct-1 accumulation. In skeletal muscle, focal disorganisation of sarcomeres was observed, while IIB-myosin heavy chain, an Oct-1 target gene, was under-expressed and rod-containing fibres were formed. These data show that a high degree of regulation of lamin B1 expression is implicated in the different clinical phenotypes observed in ADLD and show that altered Oct-1 nuclear localisation contributes to the disease phenotype.


Subject(s)
Lamin Type B/metabolism , Nuclear Envelope/metabolism , Octamer Transcription Factor-1/metabolism , Pelizaeus-Merzbacher Disease/metabolism , Blotting, Western , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cells, Cultured , Female , Fibroblasts/metabolism , Gene Duplication , Humans , Lamin Type B/genetics , Male , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Nuclear Envelope/ultrastructure , Pelizaeus-Merzbacher Disease/genetics
14.
Biochem J ; 452(2): 249-58, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23544880

ABSTRACT

GJs (gap junctions) allow direct intercellular communication, and consist of Cxs (connexins). In the mammalian central nervous system, oligodendrocytes express Cx47, Cx32 and Cx29, whereas astrocytes express Cx43, Cx30 and Cx26. Homotypic Cx47/Cx47 GJs couple oligodendrocytes, and heterotypic Cx47/Cx43 channels are the primary GJs at oligodendrocyte/astrocyte junctions. Interestingly, autosomal recessive mutations in the gene GJC2 encoding Cx47 have been linked to a central hypomyelinating disease termed PMLD (Pelizaeus-Merzbacher-like disease). The aim of the present study was to determine the cellular distribution and functional properties of PMLD-associated Cx47 mutants (I46M, G149S, G236R, G236S, M286T and T398I). Expressing GFP (green fluorescent protein)-tagged mutant versions of Cx47 in gap-junction-deficient model cells revealed that these mutants were detected at the cell-cell interface similar to that observed for wild-type Cx47. Furthermore, four of the six mutants showed no electrical coupling in both Cx47/Cx47 and Cx47/Cx43 GJ channels. These results suggest that most of the PMLD-linked Cx47 mutants disrupt Cx47/Cx47 and Cx47/Cx43 GJ function in the glial network, which may play a role in leading to PMLD symptoms.


Subject(s)
Connexin 43/genetics , Connexins/genetics , Gap Junctions/genetics , Mutant Proteins/genetics , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Base Sequence , Connexin 26 , Connexin 43/chemistry , Connexin 43/physiology , Connexins/chemistry , Connexins/metabolism , Connexins/physiology , Gap Junctions/chemistry , Gap Junctions/pathology , HeLa Cells , Humans , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/physiology , Neuroglia/chemistry , Neuroglia/metabolism , Neuroglia/pathology , Pelizaeus-Merzbacher Disease/pathology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid
15.
PLoS Genet ; 7(7): e1002146, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21750683

ABSTRACT

Gap junction channels are intercellular conduits that allow diffusional exchange of ions, second messengers, and metabolites. Human oligodendrocytes express the gap junction protein connexin47 (Cx47), which is encoded by the GJC2 gene. The autosomal recessive mutation hCx47M283T causes Pelizaeus-Merzbacher-like disease 1 (PMLD1), a progressive leukodystrophy characterized by hypomyelination, retarded motor development, nystagmus, and spasticity. We introduced the human missense mutation into the orthologous position of the mouse Gjc2 gene and inserted the mCx47M282T coding sequence into the mouse genome via homologous recombination in embryonic stem cells. Three-week-old homozygous Cx47M282T mice displayed impaired rotarod performance but unchanged open-field behavior. 10-15-day-old homozygous Cx47M282T and Cx47 null mice revealed a more than 80% reduction in the number of cells participating in glial networks after biocytin injections into oligodendrocytes in sections of corpus callosum. Homozygous expression of mCx47M282T resulted in reduced MBP expression and astrogliosis in the cerebellum of ten-day-old mice which could also be detected in Cx47 null mice of the same age. Three-month-old homozygous Cx47M282T mice exhibited neither altered open-field behavior nor impaired rotarod performance anymore. Adult mCx47M282T expressing mice did not show substantial myelin alterations, but homozygous Cx47M282T mice, additionally deprived of connexin32, which is also expressed in oligodendrocytes, died within six weeks after birth and displayed severe myelin defects accompanied by astrogliosis and activated microglia. These results strongly suggest that PMLD1 is caused by the loss of Cx47 channel function that results in impaired panglial coupling in white matter tissue.


Subject(s)
Connexins , Mutation, Missense/genetics , Oligodendroglia/metabolism , Pelizaeus-Merzbacher Disease , Animals , Connexins/deficiency , Connexins/genetics , Connexins/metabolism , Corpus Callosum/metabolism , Gap Junctions/genetics , Gap Junctions/metabolism , Humans , Ion Channels/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath/metabolism , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/metabolism , Pelizaeus-Merzbacher Disease/pathology , Stem Cells/metabolism , Gap Junction beta-1 Protein
16.
Hum Mutat ; 34(8): 1160-71, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23649844

ABSTRACT

Autosomal dominant leukodystrophy (ADLD) is an adult onset demyelinating disorder that is caused by duplications of the lamin B1 (LMNB1) gene. However, as only a few cases have been analyzed in detail, the mechanisms underlying LMNB1 duplications are unclear. We report the detailed molecular analysis of the largest collection of ADLD families studied, to date. We have identified the minimal duplicated region necessary for the disease, defined all the duplication junctions at the nucleotide level and identified the first inverted LMNB1 duplication. We have demonstrated that the duplications are not recurrent; patients with identical duplications share the same haplotype, likely inherited from a common founder and that the duplications originated from intrachromosomal events. The duplication junction sequences indicated that nonhomologous end joining or replication-based mechanisms such fork stalling and template switching or microhomology-mediated break induced repair are likely to be involved. LMNB1 expression was increased in patients' fibroblasts both at mRNA and protein levels and the three LMNB1 alleles in ADLD patients show equal expression, suggesting that regulatory regions are maintained within the rearranged segment. These results have allowed us to elucidate duplication mechanisms and provide insights into allele-specific LMNB1 expression levels.


Subject(s)
Gene Duplication , Lamin Type B/genetics , Pelizaeus-Merzbacher Disease/genetics , Adult , Base Sequence , Chromosome Breakpoints , Comparative Genomic Hybridization , DNA/chemistry , DNA/genetics , Humans , Lamin Type B/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , Pelizaeus-Merzbacher Disease/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
Biol Chem ; 394(12): 1571-81, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23959664

ABSTRACT

Myelinating cells wrap axons with multi-layered myelin sheaths for rapid impulse propagation. Dysfunctions of oligodendrocytes or Schwann cells are often associated with neuroinflammation, as observed in animal models of leukodystrophies and peripheral neuropathies, respectively. The neuroinflammatory response modulates the pathological changes, including demyelination and axonal injury, but also remyelination and repair. Here we discuss different immune mechanisms as well as factors released or exposed by myelinating glia in disease conditions. The spectrum of inflammatory mediators varies with different myelin disorders and has a major impact on the beneficial or detrimental role of immune cells in keeping nervous system integrity.


Subject(s)
Demyelinating Diseases/immunology , Neurodegenerative Diseases/immunology , Animals , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/immunology , Charcot-Marie-Tooth Disease/metabolism , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Disease Models, Animal , Inflammation/immunology , Inflammation/metabolism , Leukodystrophy, Metachromatic/genetics , Leukodystrophy, Metachromatic/immunology , Leukodystrophy, Metachromatic/metabolism , Mice , Myelin Sheath/genetics , Myelin Sheath/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Oligodendroglia/immunology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/immunology , Pelizaeus-Merzbacher Disease/metabolism , Peroxisomal Disorders/genetics , Peroxisomal Disorders/immunology , Peroxisomal Disorders/metabolism
18.
Neuroradiology ; 55(8): 933-939, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23636437

ABSTRACT

INTRODUCTION: Adult-onset ADLD with autonomic symptoms is a rare disease with a clinical course somewhat similar to chronic progressive MS but with different imaging findings consisting of extensive white matter changes in the cerebrum and cerebellar peduncles. Patients usually present in the fourth to sixth decade with autonomic symptoms, manifesting later symptoms from the pyramidal tracts and ataxia. Here, we present magnetic resonance spectroscopy (MRS) findings in this disease. METHODS: Fourteen subjects, from two non-related families, with genetic linkage to the disease were studied with magnetic resonance imaging and single-voxel MRS. Clinically, they ranged from asymptomatic to wheelchair-using. Their results were compared to those of age- and sex-matched healthy controls. RESULTS: One MRS was excluded due to suboptimal quality. The remaining 13 subjects manifested characteristic evidence of pathology on MRI, 11 of them exhibited extensive changes. The metabolite concentrations of total Cr, total Cho, and total NAA measured in millimolars, using internal water as a reference, were significantly lower in these 11 subjects compared to controls, and we found linear correlations between all these metabolite levels. When total Cr was used as a reference, we found no difference between subjects and controls. No lactate was detected. CONCLUSION: The decreased metabolite concentrations measured using internal water as a reference are most likely due to increased water content in the tissues, diluting all metabolites to a similar degree. This is also in agreement with the high signal intensity exhibited in the white matter on T2-weighted MR images and with the reported histopathological findings of vacuolated myelin.


Subject(s)
Aspartic Acid/analogs & derivatives , Autonomic Nervous System Diseases/metabolism , Brain/metabolism , Choline/metabolism , Creatine/metabolism , Magnetic Resonance Spectroscopy/methods , Pelizaeus-Merzbacher Disease/metabolism , Adult , Aged , Aspartic Acid/metabolism , Autonomic Nervous System Diseases/diagnosis , Biomarkers/metabolism , Female , Humans , Male , Middle Aged , Pelizaeus-Merzbacher Disease/diagnosis , Protons , Reproducibility of Results , Sensitivity and Specificity
19.
Biochem Biophys Res Commun ; 424(2): 262-8, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22750001

ABSTRACT

Oligodendrocytes (OLs) are myelin-forming glial cells in the central nervous system (CNS) and their dysfunction causes neuropathies such as demyelinating diseases. Proteolipid protein 1 (PLP1) is an oligodendrocyte myelin-rich tetraspan membrane protein and aberration of the plp1 gene is known to be responsible for dysmyelinating Pelizaeus-Merzbacher disease (PMD). Among previously identified gene alternations, multiplication of the plp1 gene causes increased expression of PLP1, resulting in a phenotype with severe dysmyelination in human and also rodent models. Yet little is known about the relationship between increased PLP1 expression and oligodendrocyte precursor cell (OPC) differentiation and the intracellular molecular mechanism. Here we show that expression of PLP1 in OPCs markedly inhibits their differentiation, and that this inhibitory effect is effectively improved by inhibition of extracellular signal-regulated kinase (ERK) activity. Furthermore, in cocultures with dorsal root ganglion (DRG) neurons, ERK inhibition also improves PLP1-induced dysmyelination. Thus, ERK inhibition helps to improve defective OPC differentiation induced by PLP1 expression, suggesting that molecules belonging to ERK signaling cascade may be new PMD therapeutic targets.


Subject(s)
Cell Differentiation , Extracellular Signal-Regulated MAP Kinases/metabolism , Myelin Proteolipid Protein/metabolism , Oligodendroglia/cytology , Pelizaeus-Merzbacher Disease/metabolism , Stem Cells/cytology , Coculture Techniques , Ganglia, Spinal/cytology , Humans , Myelin Sheath/metabolism , Neurons/cytology , Signal Transduction
20.
Dev Neurosci ; 34(2-3): 277-87, 2012.
Article in English | MEDLINE | ID: mdl-22832166

ABSTRACT

Pelizaeus-Merzbacher-like disease is characterized by impaired psychomotor development, ataxia, progressive spasticity and mental retardation. It is induced by mutations in the gap junction gene GJC2 that encodes for the gap junction protein connexin 47. Mice bearing a human Cx47M283T missense mutation have been generated as a transgenic mouse model of Pelizaeus-Merzbacher-like disease. Homozygous expression of the mutant connexin 47 gene in oligodendrocytes resulted in a complex and variable neuropathologic phenotype, which was associated with impairments in motor coordination in juvenile, but not adult mice. In the present study, we have investigated anxiety-like behaviour and spatial working memory in juvenile (P23) and adult (3-month-old) Cx47M282T mutant mice. Adult Cx47M282T mice were also evaluated in terms of neuromotor functions and in the novel object recognition test. Juvenile Cx47M282T mutant mice exhibited an increase in anxiety-like behaviour in the open field test, but no changes in spatial working memory performance. No significant changes in anxiety-like behaviour, spatial working memory or neuromotor functions were observed in the adult Cx47M282T mutant mice. However, novel object recognition was significantly impaired in adult Cx47M282T mice. Our results suggest that the expression of the human Cx47M282T mutation in the mouse causes changes in anxiety-like behaviour in juvenile and novel object recognition impairments in adult mice. It appears that the distortion of panglial gap junction coupling in white and grey matter tissue in the Cx47M282T mice is associated with a complex age-dependent behavioural phenotype including changes in psychomotor, emotional and memory functions.


Subject(s)
Anxiety/genetics , Behavior, Animal/physiology , Connexins/genetics , Recognition, Psychology/physiology , Age Factors , Animals , Anxiety/metabolism , Connexins/metabolism , Disease Models, Animal , Gap Junctions/genetics , Gap Junctions/metabolism , Memory, Short-Term/physiology , Mice , Mutation , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL