Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Stem Cell Reports ; 11(3): 711-726, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30146490

ABSTRACT

Pelizaeus-Merzbacher disease (PMD) is a fatal X-linked disorder caused by loss of myelinating oligodendrocytes and consequent hypomyelination. The underlying cellular and molecular dysfunctions are not fully defined, but therapeutic enhancement of oligodendrocyte survival could restore functional myelination in patients. Here we generated pure, scalable quantities of induced pluripotent stem cell-derived oligodendrocyte progenitor cells (OPCs) from a severe mouse model of PMD, Plp1jimpy. Temporal phenotypic and transcriptomic studies defined an early pathological window characterized by endoplasmic reticulum (ER) stress and cell death as OPCs exit their progenitor state. High-throughput phenotypic screening identified a compound, Ro 25-6981, which modulates the ER stress response and rescues mutant oligodendrocyte survival in jimpy, in vitro and in vivo, and in human PMD oligocortical spheroids. Surprisingly, increasing oligodendrocyte survival did not restore subsequent myelination, revealing a second pathological phase. Collectively, our work shows that PMD oligodendrocyte loss can be rescued pharmacologically and defines a need for multifactorial intervention to restore myelination.


Subject(s)
Oligodendrocyte Precursor Cells/pathology , Pelizaeus-Merzbacher Disease/pathology , Animals , Cell Survival , Cells, Cultured , Disease Models, Animal , Endoplasmic Reticulum Stress , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mice , Mutation , Myelin Sheath/genetics , Myelin Sheath/metabolism , Myelin Sheath/pathology , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/metabolism , Transcriptome
8.
Neurosci Lett ; 678: 90-98, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29729355

ABSTRACT

Proteolipid protein (PLP), besides its adhesive role in myelin, has been postulated to have multiple cellular functions. One well-documented function of PLP is regulation of oligodendrocyte (Olg) apoptosis. In contrast, DM20, an alternatively spliced product of the PLP1/Plp1 gene, has been proposed to have functions that are unique from PLP but these functions have never been elucidated. Here, we compare metabolism of PLP and DM20, and show that oxidative phosphorylation (OxPhos) was significantly decreased in Plp1 but not DM20 or EGFP expressing cells. The reserve OxPhos capacity of Plp1 expressing cells was half of control cells, suggesting that they are very vulnerable to stress. ATP in media of Plp1 expressing cells is significantly increased more than two-fold compared to controls; markers of apoptosis are increased in cells over-expressing Plp1, indicating that abnormal metabolism of PLP is most likely the direct cause leading to Olg apoptosis. We hypothesize that abnormal metabolism, mediated by increased insertion of PLP into mitochondria, underlies demyelination in Pelizaeus-Merzbacher Disease (PMD) and in models of PMD. To understand why PLP and DM20 function differently, we mutated or deleted amino acids located in the PLP-specific region. All these mutations and deletions of the PLP-specific region prevented insertion of PLP into mitochondria. These findings demonstrate that the PLP-specific region is essential for PLP's import into mitochondria, and now offer an explanation for deciphering unique functions of PLP and DM20.


Subject(s)
Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Myelin Proteolipid Protein/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis , COS Cells , Cell Respiration , Chlorocebus aethiops , Lactic Acid/metabolism , Pelizaeus-Merzbacher Disease/metabolism
9.
Brain Pathol ; 28(5): 611-630, 2018 09.
Article in English | MEDLINE | ID: mdl-29027761

ABSTRACT

Pelizaeus-Merzbacher disease (PMD) is a fatal hypomyelinating disorder characterized by early impairment of motor development, nystagmus, choreoathetotic movements, ataxia and progressive spasticity. PMD is caused by variations in the proteolipid protein gene PLP1, which encodes the two major myelin proteins of the central nervous system, PLP and its spliced isoform DM20, in oligodendrocytes. Large duplications including the entire PLP1 gene are the most frequent causative mutation leading to the classical form of PMD. The Plp1 overexpressing mouse model (PLP-tg66/66 ) develops a phenotype very similar to human PMD, with early and severe motor dysfunction and a dramatic decrease in lifespan. The sequence of cellular events that cause neurodegeneration and ultimately death is poorly understood. In this work, we analyzed patient-derived fibroblasts and spinal cords of the PLP-tg66/66 mouse model, and identified redox imbalance, with altered antioxidant defense and oxidative damage to several enzymes involved in ATP production, such as glycolytic enzymes, creatine kinase and mitochondrial proteins from the Krebs cycle and oxidative phosphorylation. We also evidenced malfunction of the mitochondria compartment with increased ROS production and depolarization in PMD patient's fibroblasts, which was prevented by the antioxidant N-acetyl-cysteine. Finally, we uncovered an impairment of mitochondrial dynamics in patient's fibroblasts which may help explain the ultrastructural abnormalities of mitochondria morphology detected in spinal cords from PLP-tg66/66 mice. Altogether, these results underscore the link between redox and metabolic homeostasis in myelin diseases, provide insight into the pathophysiology of PMD, and may bear implications for tailored pharmacological intervention.


Subject(s)
Mitochondrial Dynamics , Oxidative Stress , Pelizaeus-Merzbacher Disease/metabolism , Animals , Cells, Cultured , Child , Child, Preschool , DNA, Mitochondrial , Fibroblasts/metabolism , Fibroblasts/pathology , Glutamic Acid/metabolism , Humans , Infant , Male , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Pelizaeus-Merzbacher Disease/pathology , RNA, Messenger/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology
11.
Nucleus ; 7(6): 547-553, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27854160

ABSTRACT

Autosomal Dominant Leukodystrophy (ADLD), a fatal adult onset demyelinating disorder, is the only human disease that has been linked to mutations of the nuclear lamina protein, lamin B1, and is primarily caused by duplications of the LMNB1 gene. Why CNS myelin is specifically targeted and the mechanisms underlying ADLD are unclear. Recent work from our group has demonstrated that over expression of lamin B1 in oligodendrocytes, the myelin producing cells in the CNS, resulted in age dependent epigenetic modifications, transcriptional down-regulation of lipogenic gene expression and significant reductions of myelin-enriched lipids. Given the high lipid content of meylin, we hypothesize that lipid loss is one of the primary drivers of the demyelination phenotype. These results can, at least partially, explain the age dependence and cell type specificity in ADLD and are discussed in the context of the existing literature, in an attempt to delineate potential pathways underlying the disease phenotype.


Subject(s)
Lamin Type B/metabolism , Lipid Metabolism , Pelizaeus-Merzbacher Disease/metabolism , Animals , Epigenesis, Genetic , Humans , Lamin Type B/genetics , Mutation , Pelizaeus-Merzbacher Disease/genetics , Transcription, Genetic
12.
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
13.
Brain Dev ; 38(6): 581-4, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26725305

ABSTRACT

A patient with an unusually mild form of Pelizaeus-Merzbacher disease was studied. Clinically, mild developmental delay with acquisition of assisted walking at 16months and mild spastic tetraplegia were evident, but no nystagmus, cerebellar, or extra-pyramidal signs were present. PLP1 mutation analysis revealed a nucleotide substitution adjacent to the acceptor site of intron 3, NM_000533.4:c.454-9T>G. Expression analysis using the patient's leukocytes demonstrated an additional abnormal transcript including the last 118bp of intron 3. In silico prediction analysis suggested the reduction of wild-type acceptor activity, which presumably evokes the cryptic splicing variant. Putative cryptic transcript results in premature termination, which may explain the mild clinical phenotype observed in this patient.


Subject(s)
Mutation , Myelin Proteolipid Protein/genetics , Pelizaeus-Merzbacher Disease/genetics , Brain/diagnostic imaging , Child , DNA Mutational Analysis , Humans , Introns , Leukocytes/metabolism , Magnetic Resonance Imaging , Male , Myelin Proteolipid Protein/metabolism , Pelizaeus-Merzbacher Disease/diagnostic imaging , Pelizaeus-Merzbacher Disease/metabolism , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Severity of Illness Index
14.
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
15.
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
16.
Brain Dev ; 37(8): 797-802, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25491635

ABSTRACT

PURPOSE: Pelizaeus-Merzbacher disease (PMD) is a rare X-linked recessive hypomyelination disorder characterized by nystagmus, ataxia, impaired motor development, and progressive spasticity. Identification of proteolipid protein 1 (PLP1) mutations in Chinese patients with Pelizaeus-Merzbacher disease (PMD) and confirmation of the biological impacts of the identified mutations are the aims of this study. METHODS: An analysis of clinical materials and a follow-up study were conducted for the patients with PMD. Sequencing and immunofluorescence were applied for molecular analysis of the causative gene PLP1. RESULTS: We identified PLP1 mutations in seven male patients with PMD. Three novel missense mutations (c.353C>G, p.T118R; c.623G>T, p.G208V; c.709T>G, p.F237V) and three reported missense mutations (c.467C>T, p.T156I; c.517C>T, p.P173S; c.646C>T, p.P216S) of PLP1 were identified from seven Chinese PMD patients. The three mutations (F237V in patient 2, P216S in patient 5 and T156I in patient 6) were de novo. Mutant proteins were trapped in the lumen of endoplasmic reticulum. CONCLUSION: We have identified six pathogenic mutations, enriching the specific spectrum of missense mutations in the patients with PMD. The six PLP1 mutations are probably pathogenic. By reviewing the known PLP1 mutations, we have preliminarily revealed the position of missense mutation may be associated with the severity of PMD.


Subject(s)
Asian People/genetics , Mutation, Missense , Myelin Proteolipid Protein/genetics , Pelizaeus-Merzbacher Disease/genetics , Child , Child, Preschool , China , Genetic Association Studies , Humans , Infant , Male , Myelin Proteolipid Protein/metabolism , Pelizaeus-Merzbacher Disease/metabolism , Pelizaeus-Merzbacher Disease/pathology
17.
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
18.
J Child Neurol ; 29(2): 283-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24056155

ABSTRACT

A boy with Pelizaeus-Merzbacher disease underwent repeated evaluations by 3-Tesla (1)H-magnetic resonance spectroscopy (MRS). The patient showed overlap of the PLP1. Individuals selected as normal controls for (1)H-magnetic resonance spectroscopy consisted of healthy age-matched children. For (1)H-magnetic resonance spectroscopy, the center of a voxel was positioned in the right parietal lobe. (1)H-magnetic resonance spectroscopy was performed when the patient was 2, 6, 14, and 25 months old. γ-Aminobutyric acid concentration in early childhood was increased compared with that in normal controls. However, the γ-aminobutyric acid concentration in the Pelizaeus-Merzbacher disease patient was normalized at 14 and 25 months. No remarkable changes were observed in choline-containing compounds concentration at any time. These results suggest that the changes in metabolite concentrations during growth can reflect the pathological condition of Pelizaeus-Merzbacher disease. Furthermore, the lack of change in the choline-containing compounds concentration can be useful for differentiating Pelizaeus-Merzbacher disease from other white matter disorders.


Subject(s)
Parietal Lobe/growth & development , Parietal Lobe/metabolism , Pelizaeus-Merzbacher Disease/metabolism , Aging , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Case-Control Studies , Child, Preschool , Creatine/metabolism , Glutamic Acid/metabolism , Humans , Infant , Inositol/metabolism , Magnetic Resonance Spectroscopy , Male , gamma-Aminobutyric Acid/metabolism
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...