Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 316
Filter
1.
ACS Appl Mater Interfaces ; 14(38): 42887-42903, 2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36094079

ABSTRACT

Acetaminophen (APAP)-induced liver injury (AILI) is a common liver disease in clinical practice. Only one clinically approved drug, N-acetylcysteine (NAC), for the treatment of AILI is available in clinics, but novel treatment strategies are still needed due to the complicated pathological changes of AILI and the side effects of NAC. Here, we found that astaxanthin (ASX) can prevent AILI through the Nrf2/HO-1 pathway. After treatment with ASX, there was a positive activation of the Nrf2/HO-1 pathway in AILI models both in vivo and in vitro accompanied by enhanced autophagy and reduced ferroptosis. In APAP-challenged L02 liver cells, ASX reduced autophagy and enhanced apoptosis of the cells. Furthermore, we developed ASX-loaded hollow mesoporous silica nanoparticles (HMSN@ASX) to improve the aqueous solubility of ASX and targeted delivery of ASX to the liver and then significantly improve the therapeutic effects. Taken together, we found that ASX can protect against AILI by activating the Nrf2/HO-1 pathway, which mainly affects oxidative stress, autophagy, and ferroptosis processes, and the HMSN@ASX nanosystem can target the liver to enhance the treatment efficiency of AILI.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Chemical and Drug Induced Liver Injury , Ferroptosis , Hereditary Sensory and Motor Neuropathy , Acetaminophen/metabolism , Acetylcysteine , Autophagy , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury, Chronic/drug therapy , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Chemical and Drug Induced Liver Injury, Chronic/pathology , Heme Oxygenase-1/metabolism , Hereditary Sensory and Motor Neuropathy/drug therapy , Hereditary Sensory and Motor Neuropathy/metabolism , Hereditary Sensory and Motor Neuropathy/pathology , Humans , Liver/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Silicon Dioxide/pharmacology , Xanthophylls
2.
Neuromuscul Disord ; 32(6): 503-511, 2022 06.
Article in English | MEDLINE | ID: mdl-35501275

ABSTRACT

Focal thickening of the myelin sheath, also known as tomacula, is a characteristic pathological feature of patients with hereditary neuropathy with liability to pressure palsies (HNPP). However, a deeper understanding of the pathology underlying unmyelinated fibers and nonmyelinating Schwann cells is required. Electron microscopic examination of sural nerve biopsy specimens was performed for 14 HNPP patients with peripheral myelin protein 22 (PMP22) deletion, and their results were compared to 12 normal controls and 14 Charcot-Marie-Tooth disease type 1A (CMT1A) patients with PMP22 duplication. The number of unmyelinated axons in a single axon-containing nonmyelinating Schwann cell subunit in the HNPP group significantly increased compared with that in normal controls (1.99 ±â€¯0.66 vs. 1.57 ±â€¯0.52, p < 0.05). Conversely, these numbers significantly decreased in the CMT1A group compared with those in normal controls (1.16 ±â€¯0.16, p < 0.05). Some unmyelinated axons in patients with HNPP were incompletely surrounded by the cytoplasm of Schwann cells, almost as if the Schwann cells failed to form mesaxons; such failure in mesaxon formation was not observed in normal controls or in patients with CMT1A. These findings suggest that PMP22 dosage affects nonmyelinating as well as myelinating Schwann cells.


Subject(s)
Charcot-Marie-Tooth Disease , Hereditary Sensory and Motor Neuropathy , Arthrogryposis , Charcot-Marie-Tooth Disease/diagnosis , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/pathology , Humans , Myelin Proteins/genetics , Myelin Proteins/metabolism , Schwann Cells/pathology
3.
Biomolecules ; 11(8)2021 07 28.
Article in English | MEDLINE | ID: mdl-34439778

ABSTRACT

Neuro-muscular disorders include a variety of diseases induced by genetic mutations resulting in muscle weakness and waste, swallowing and breathing difficulties. However, muscle alterations and nerve depletions involve specific molecular and cellular mechanisms which lead to the loss of motor-nerve or skeletal-muscle function, often due to an excessive cell death. Morphological and molecular studies demonstrated that a high number of these disorders seem characterized by an upregulated apoptosis which significantly contributes to the pathology. Cell death involvement is the consequence of some cellular processes that occur during diseases, including mitochondrial dysfunction, protein aggregation, free radical generation, excitotoxicity and inflammation. The latter represents an important mediator of disease progression, which, in the central nervous system, is known as neuroinflammation, characterized by reactive microglia and astroglia, as well the infiltration of peripheral monocytes and lymphocytes. Some of the mechanisms underlying inflammation have been linked to reactive oxygen species accumulation, which trigger mitochondrial genomic and respiratory chain instability, autophagy impairment and finally neuron or muscle cell death. This review discusses the main inflammatory pathways contributing to cell death in neuro-muscular disorders by highlighting the main mechanisms, the knowledge of which appears essential in developing therapeutic strategies to prevent the consequent neuron loss and muscle wasting.


Subject(s)
Apoptosis/genetics , Hereditary Sensory and Motor Neuropathy/metabolism , Motor Neuron Disease/metabolism , Muscular Diseases/metabolism , Muscular Dystrophies/metabolism , Neuromuscular Junction Diseases/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Autophagy/genetics , Cytokines/genetics , Cytokines/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/pathology , Humans , Inflammation , Microglia/metabolism , Microglia/pathology , Mitochondria/metabolism , Mitochondria/pathology , Motor Neuron Disease/genetics , Motor Neuron Disease/pathology , Muscular Diseases/genetics , Muscular Diseases/pathology , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Neuromuscular Junction Diseases/genetics , Neuromuscular Junction Diseases/pathology , Neurons/metabolism , Neurons/pathology , Signal Transduction
4.
Ann Clin Transl Neurol ; 8(6): 1330-1342, 2021 06.
Article in English | MEDLINE | ID: mdl-33943039

ABSTRACT

BACKGROUND: The expansion of GGC repeat in the 5' untranslated region of the NOTCH2NLC has been associated with various neurogenerative disorders of the central nervous system and, more recently, oculopharyngodistal myopathy. This study aimed to report patients with distal weakness with both neuropathic and myopathic features on electrophysiology and pathology who present GGC repeat expansions in the NOTCH2NLC. METHODS: Whole-exome sequencing (WES) and long-read sequencing were implemented to identify the candidate genes. In addition, the available clinical data and the pathological changes associated with peripheral nerve and muscle biopsies were reviewed and studied. RESULTS: We identified and validated GGC repeat expansions of NOTCH2NLC in three unrelated patients who presented with progressive weakness predominantly affecting distal lower limb muscles, following negative results in an initial WES. We found intranuclear inclusions with multiple proteins deposits in the nuclei of both myofibers and Schwann cells. The clinical features of these patients are compatible with the diagnosis of distal motor neuropathy and rimmed vacuolar myopathy. INTERPRETATION: These phenotypes enrich the class of features associated with NOTCH2NLC-related repeat expansion disorders (NRED), and provide further evidence that the neurological symptoms of NRED include not only brain, spinal cord, and peripheral nerves damage, but also myopathy, and that overlapping symptoms might exist.


Subject(s)
Distal Myopathies/genetics , Hereditary Sensory and Motor Neuropathy/genetics , Intercellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/genetics , Adult , Distal Myopathies/pathology , Distal Myopathies/physiopathology , Female , Hereditary Sensory and Motor Neuropathy/pathology , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Intranuclear Inclusion Bodies/genetics , Male , Middle Aged , Pedigree , Phenotype , Trinucleotide Repeat Expansion/genetics , Exome Sequencing
5.
J Mol Neurosci ; 71(12): 2462-2467, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33791913

ABSTRACT

Sensory ataxic neuropathy, dysarthria, and ophthalmoparesis (SANDO) is a rare mitochondrial disorder associated with mutations in the POLG gene, which encodes the DNA polymerase gamma catalytic subunit. A few POLG-related SANDO cases have been reported, but the genotype-phenotype correlation remains unclear. Here, we report a patient with SANDO carrying two novel missense variants (c.2543G>C, p.G848A and c.452 T>C, p.L151P) in POLG. We also reviewed previously reported cases to systematically evaluate the clinical and genetic features of POLG-related SANDO. A total of 35 distinct variants in the coding region of POLG were identified in 63 patients with SANDO. The most frequent variant was the p.A467T variant, followed by the p.W748S variant. The clinical spectrum of SANDO is heterogeneous. No clear correlation has been observed between the mutation types and clinical phenotypes. Our findings expand the mutational spectrum of POLG and contribute to clinical management and genetic counseling for POLG-related SANDO.


Subject(s)
DNA Polymerase gamma/genetics , Dysarthria/genetics , Hereditary Sensory and Motor Neuropathy/genetics , Ophthalmoplegia/genetics , Adult , Dysarthria/pathology , Hereditary Sensory and Motor Neuropathy/pathology , Humans , Male , Mutation, Missense , Ophthalmoplegia/pathology , Phenotype
6.
Brain ; 144(2): 584-600, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33559681

ABSTRACT

The extracellular matrix comprises a network of macromolecules such as collagens, proteoglycans and glycoproteins. VWA1 (von Willebrand factor A domain containing 1) encodes a component of the extracellular matrix that interacts with perlecan/collagen VI, appears to be involved in stabilizing extracellular matrix structures, and demonstrates high expression levels in tibial nerve. Vwa1-deficient mice manifest with abnormal peripheral nerve structure/function; however, VWA1 variants have not previously been associated with human disease. By interrogating the genome sequences of 74 180 individuals from the 100K Genomes Project in combination with international gene-matching efforts and targeted sequencing, we identified 17 individuals from 15 families with an autosomal-recessive, non-length dependent, hereditary motor neuropathy and rare biallelic variants in VWA1. A single disease-associated allele p.(G25Rfs*74), a 10-bp repeat expansion, was observed in 14/15 families and was homozygous in 10/15. Given an allele frequency in European populations approaching 1/1000, the seven unrelated homozygote individuals ascertained from the 100K Genomes Project represents a substantial enrichment above expected. Haplotype analysis identified a shared 220 kb region suggesting that this founder mutation arose >7000 years ago. A wide age-range of patients (6-83 years) helped delineate the clinical phenotype over time. The commonest disease presentation in the cohort was an early-onset (mean 2.0 ± 1.4 years) non-length-dependent axonal hereditary motor neuropathy, confirmed on electrophysiology, which will have to be differentiated from other predominantly or pure motor neuropathies and neuronopathies. Because of slow disease progression, ambulation was largely preserved. Neurophysiology, muscle histopathology, and muscle MRI findings typically revealed clear neurogenic changes with single isolated cases displaying additional myopathic process. We speculate that a few findings of myopathic changes might be secondary to chronic denervation rather than indicating an additional myopathic disease process. Duplex reverse transcription polymerase chain reaction and immunoblotting using patient fibroblasts revealed that the founder allele results in partial nonsense mediated decay and an absence of detectable protein. CRISPR and morpholino vwa1 modelling in zebrafish demonstrated reductions in motor neuron axonal growth, synaptic formation in the skeletal muscles and locomotive behaviour. In summary, we estimate that biallelic variants in VWA1 may be responsible for up to 1% of unexplained hereditary motor neuropathy cases in Europeans. The detailed clinical characterization provided here will facilitate targeted testing on suitable patient cohorts. This novel disease gene may have previously evaded detection because of high GC content, consequential low coverage and computational difficulties associated with robustly detecting repeat-expansions. Reviewing previously unsolved exomes using lower QC filters may generate further diagnoses.


Subject(s)
Extracellular Matrix Proteins/genetics , Hereditary Sensory and Motor Neuropathy/genetics , Adult , Aged , Animals , Behavior, Animal/physiology , Child , Female , Hereditary Sensory and Motor Neuropathy/pathology , Humans , Male , Middle Aged , Muscle, Skeletal/pathology , Mutation , Pedigree , Young Adult , Zebrafish
7.
Annu Rev Pathol ; 16: 487-509, 2021 01 24.
Article in English | MEDLINE | ID: mdl-33497257

ABSTRACT

Hereditary peripheral neuropathy (HPN) is a complex group of neurological disorders caused by mutations in genes expressed by neurons and Schwann cells. The inheritance of a single mutation or multiple mutations in several genes leads to disease phenotype. Patients exhibit symptoms during development, at an early age or later in adulthood. Most of the mechanistic understanding about these neuropathies comes from animal models and histopathological analyses of postmortem human tissues. Diagnosis is often very complex due to the heterogeneity and overlap in symptoms and the frequent overlap between various genes and different mutations they possess. Some symptoms in HPN are common through different subtypes such as axonal degeneration, demyelination, and loss of motor and sensory neurons, leading to similar physiologic abnormalities. Recent advances in gene-targeted therapies, genetic engineering, and next-generation sequencing have augmented our understanding of the underlying pathogenetic mechanisms of HPN.


Subject(s)
Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/pathology , Adult , Animals , Hereditary Sensory and Motor Neuropathy/diagnosis , Humans , Mutation
8.
Neurosci Lett ; 744: 135595, 2021 01 23.
Article in English | MEDLINE | ID: mdl-33359733

ABSTRACT

Development of peripheral nervous system (PNS) myelin involves a coordinated series of events between growing axons and the Schwann cell (SC) progenitors that will eventually ensheath them. Myelin sheaths have evolved out of necessity to maintain rapid impulse propagation while accounting for body space constraints. However, myelinating SCs perform additional critical functions that are required to preserve axonal integrity including mitigating energy consumption by establishing the nodal architecture, regulating axon caliber by organizing axonal cytoskeleton networks, providing trophic and potentially metabolic support, possibly supplying genetic translation materials and protecting axons from toxic insults. The intermediate steps between the loss of these functions and the initiation of axon degeneration are unknown but the importance of these processes provides insightful clues. Prevalent demyelinating diseases of the PNS include the inherited neuropathies Charcot-Marie-Tooth Disease, Type 1 (CMT1) and Hereditary Neuropathy with Liability to Pressure Palsies (HNPP) and the inflammatory diseases Acute Inflammatory Demyelinating Polyneuropathy (AIDP) and Chronic Inflammatory Demyelinating Polyneuropathy (CIDP). Secondary axon degeneration is a common feature of demyelinating neuropathies and this process is often correlated with clinical deficits and long-lasting disability in patients. There is abundant electrophysiological and histological evidence for secondary axon degeneration in patients and rodent models of PNS demyelinating diseases. Fully understanding the involvement of secondary axon degeneration in these diseases is essential for expanding our knowledge of disease pathogenesis and prognosis, which will be essential for developing novel therapeutic strategies.


Subject(s)
Axons/metabolism , Demyelinating Diseases/metabolism , Nerve Degeneration/metabolism , Polyneuropathies/metabolism , Animals , Arthrogryposis/metabolism , Arthrogryposis/pathology , Axons/pathology , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Demyelinating Diseases/pathology , Hereditary Sensory and Motor Neuropathy/metabolism , Hereditary Sensory and Motor Neuropathy/pathology , Humans , Nerve Degeneration/pathology , Polyneuropathies/pathology , Schwann Cells/metabolism , Schwann Cells/pathology
9.
Int J Mol Sci ; 21(20)2020 Oct 15.
Article in English | MEDLINE | ID: mdl-33076433

ABSTRACT

Diseases associated with acquired or genetic defects in members of the chaperoning system (CS) are increasingly found and have been collectively termed chaperonopathies. Illustrative instances of genetic chaperonopathies involve the genes for chaperonins of Groups I (e.g., Heat shock protein 60, Hsp60) and II (e.g., Chaperonin Containing T-Complex polypeptide 1, CCT). Examples of the former are hypomyelinating leukodystrophy 4 (HLD4 or MitCHAP60) and hereditary spastic paraplegia (SPG13). A distal sensory mutilating neuropathy has been linked to a mutation [p.(His147Arg)] in subunit 5 of the CCT5 gene. Here, we describe a new possibly pathogenic variant [p.(Leu224Val)] of the same subunit but with a different phenotype. This yet undescribed disease affects a girl with early onset demyelinating neuropathy and a severe motor disability. By whole exome sequencing (WES), we identified a homozygous CCT5 c.670C>G p.(Leu224Val) variant in the CCT5 gene. In silico 3D-structure analysis and bioinformatics indicated that this variant could undergo abnormal conformation and could be pathogenic. We compared the patient's clinical, neurophysiological and laboratory data with those from patients carrying p.(His147Arg) in the equatorial domain. Our patient presented signs and symptoms absent in the p.(His147Arg) cases. Molecular dynamics simulation and modelling showed that the Leu224Val mutation that occurs in the CCT5 intermediate domain near the apical domain induces a conformational change in the latter. Noteworthy is the striking difference between the phenotypes putatively linked to mutations in the same CCT subunit but located in different structural domains, offering a unique opportunity for elucidating their distinctive roles in health and disease.


Subject(s)
Chaperonin Containing TCP-1/genetics , Hereditary Sensory and Motor Neuropathy/genetics , Mutation, Missense , Age of Onset , Chaperonin Containing TCP-1/chemistry , Female , Hereditary Sensory and Motor Neuropathy/pathology , Humans , Infant, Newborn , Molecular Dynamics Simulation , Myelin Sheath/metabolism , Phenotype
10.
J Peripher Nerv Syst ; 25(4): 423-428, 2020 12.
Article in English | MEDLINE | ID: mdl-33107133

ABSTRACT

Minifascicular neuropathy (MN) is a rare, autosomal recessive disease with prominent structural changes of peripheral nerves. So far, it has been observed in females with a 46,XY karyotype and mutations of the Desert Hedgehog (DHH) gene, thus linking MN to gonadal dysgenesis (GD) and disorders of sex development (DSD). However, a 46,XX proband with normal female sex and gender development underwent clinical evaluations, nerve conduction studies and genetic screening for a severe motor-sensory neuropathy with a pathological phenotype that hinted at MN. Indeed, sural nerve biopsy revealed a profound disturbance of perineurium development with a thin and loose structure. High-resolution ultrasound (HRUS) also disclosed diffuse changes of nerve echotexture that visibly correlated with the pathological features. After extensive genetic testing, a novel homozygous DHH null mutation (p.Ser185*) was identified in the proband and in her sister, who was affected by a similar motor-sensory neuropathy, but was eventually found to be a 46,XY patient according to a late diagnosis of DSD with complete GD. DHH should therefore be considered as a possible cause of rare non-syndromic hereditary motor-sensory neuropathies, regardless of DSD. Furthermore, HRUS could effectively smooth the complex diagnostic workup as it demonstrated a high predictive power to detect MN, providing the same detailed correlations to the pathologic features of the nerve biopsy and Dhh-/- mice in both sisters. Hence, HRUS may assume a pivotal role in guiding molecular analysis in individuals with or without DSD.


Subject(s)
Disorder of Sex Development, 46,XY/diagnosis , Hedgehog Proteins/genetics , Hereditary Sensory and Motor Neuropathy , Consanguinity , Female , Genetic Testing , Hereditary Sensory and Motor Neuropathy/diagnosis , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/pathology , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Microscopy, Acoustic , Middle Aged , Siblings , Sural Nerve/pathology , Syndrome
12.
Hum Mol Genet ; 29(10): 1689-1699, 2020 06 27.
Article in English | MEDLINE | ID: mdl-32356557

ABSTRACT

Copy number variation of the peripheral nerve myelin gene Peripheral Myelin Protein 22 (PMP22) causes multiple forms of inherited peripheral neuropathy. The duplication of a 1.4 Mb segment surrounding this gene in chromosome 17p12 (c17p12) causes the most common form of Charcot-Marie-Tooth disease type 1A, whereas the reciprocal deletion of this gene causes a separate neuropathy termed hereditary neuropathy with liability to pressure palsies (HNPP). PMP22 is robustly induced in Schwann cells in early postnatal development, and several transcription factors and their cognate regulatory elements have been implicated in coordinating the gene's proper expression. We previously found that a distal super-enhancer domain was important for Pmp22 expression in vitro, with particular impact on a Schwann cell-specific alternative promoter. Here, we investigate the consequences of deleting this super-enhancer in vivo. We find that loss of the super-enhancer in mice reduces Pmp22 expression throughout development and into adulthood, with greater impact on the Schwann cell-specific promoter. Additionally, these mice display tomacula formed by excessive myelin folding, a pathological hallmark of HNPP, as have been previously observed in heterozygous Pmp22 mice as well as sural biopsies from patients with HNPP. Our findings demonstrate a mechanism by which smaller copy number variations, not including the Pmp22 gene, are sufficient to reduce gene expression and phenocopy a peripheral neuropathy caused by the HNPP-associated deletion encompassing PMP22.


Subject(s)
Arthrogryposis/genetics , Charcot-Marie-Tooth Disease/genetics , Enhancer Elements, Genetic/genetics , Hereditary Sensory and Motor Neuropathy/genetics , Myelin Proteins/genetics , Adult , Animals , Arthrogryposis/metabolism , Arthrogryposis/pathology , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , DNA Copy Number Variations/genetics , Hereditary Sensory and Motor Neuropathy/metabolism , Hereditary Sensory and Motor Neuropathy/pathology , Heterozygote , Humans , Mice , Myelin Sheath/genetics , Myelin Sheath/metabolism , Peripheral Nerves/metabolism , Peripheral Nerves/pathology , Phenotype , Schwann Cells/metabolism , Schwann Cells/pathology
13.
Neuromuscul Disord ; 30(3): 227-231, 2020 03.
Article in English | MEDLINE | ID: mdl-32085962

ABSTRACT

The Tropomyosin-receptor kinase fused gene (TFG) encodes TFG which is expressed in spinal motor neurons, dorsal root ganglia and cranial nerve nuclei, and plays a role in the dynamics of the endoplasmic reticulum. Two dominant missense TFG mutations have previously been reported in limited geographical areas (Far East, Iran, China) in association with hereditary motor sensory neuropathy with proximal involvement (HMSN-P) of the four limbs, or with Charcot-Marie-Tooth disease type 2 (CMT2). The 60-year-old female proband belonging to a three-generation Italian family presented with an atypical neuropathy characterized by diffuse painful cramps and prominent motor-sensory impairment of the distal upper limbs. Her sural nerve biopsy showed chronic axonal neuropathy without active degeneration or regeneration. Targeted next-generation sequencing of a panel with 98 genes associated with inherited peripheral neuropathies/neuromuscular disorders identified three candidate genes: TFG, DHTKD1 and DCTN2. In the family, the disease co-segregated with the TFG p.(Gly269Val) variant. TFG should be considered in genetic testing of patients with heterogeneous inherited neuropathy, independently of their ethnic origin.


Subject(s)
Hereditary Sensory and Motor Neuropathy , Proteins/genetics , Upper Extremity/physiopathology , Female , Hereditary Sensory and Motor Neuropathy/diagnosis , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/pathology , Hereditary Sensory and Motor Neuropathy/physiopathology , High-Throughput Nucleotide Sequencing , Humans , Middle Aged , Pedigree
14.
Ann Clin Transl Neurol ; 7(1): 15-25, 2020 01.
Article in English | MEDLINE | ID: mdl-31872979

ABSTRACT

OBJECTIVE: Hereditary neuropathy with liability to pressure palsies (HNPP) is caused by heterozygous deletion of the peripheral myelin protein 22 (PMP22) gene. Patients with HNPP present multifocal, reversible sensory/motor deficits due to increased susceptibility to mechanical pressure. Additionally, age-dependent axonal degeneration is reported. We hypothesize that length-dependent axonal loss can be revealed by MRI, irrespective of the multifocal phenotype in HNPP. METHODS: Nerve and muscle MRI data were acquired in the proximal and distal leg of patients with HNPP (n = 10) and matched controls (n = 7). More specifically, nerve magnetization transfer ratios (MTR) were evaluated to assay proximal-to-distal gradients in nerve degeneration, while intramuscular fat percentages (Fper ) were evaluated to assay muscle fat replacement following denervation. Neurological disabilities were assessed via the Charcot-Marie-Tooth neuropathy score (CMTNS) for correlation with MRI. RESULTS: Fper values were elevated in HNPP proximal muscle (9.8 ± 2.2%, P = 0.01) compared to controls (6.9 ± 1.0%). We observed this same elevation of HNPP distal muscles (10.5 ± 2.5%, P < 0.01) relative to controls (6.3 ± 1.1%). Additionally, the amplitude of the proximal-to-distal gradient in Fper was more significant in HNPP patients than controls (P < 0.01), suggesting length-dependent axonal loss. In contrast, nerve MTR values were similar between HNPP subjects (sciatic/tibial nerves = 39.4 ± 2.0/34.2 ± 2.5%) and controls (sciatic/tibial nerves = 37.6 ± 3.8/35.5 ± 1.2%). Proximal muscle Fper values were related to CMTNS (r = 0.69, P = 0.03), while distal muscle Fper and sciatic/tibial nerve MTR values were not related to disability. INTERPRETATION: Despite the multifocal nature of the HNPP phenotype, muscle Fper measurements relate to disability and exhibit a proximal-to-distal gradient consistent with length-dependent axonal loss, suggesting that Fper may be a viable biomarker of disease progression in HNPP.


Subject(s)
Adiposity , Arthrogryposis/diagnostic imaging , Axons/pathology , Hereditary Sensory and Motor Neuropathy/diagnostic imaging , Leg/diagnostic imaging , Muscle, Skeletal/diagnostic imaging , Nerve Degeneration/diagnostic imaging , Sciatic Nerve/diagnostic imaging , Adolescent , Adult , Arthrogryposis/pathology , Female , Hereditary Sensory and Motor Neuropathy/pathology , Humans , Leg/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Nerve Degeneration/pathology , Sciatic Nerve/pathology , Young Adult
15.
Eur J Med Genet ; 63(4): 103826, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31857255

ABSTRACT

Mutations in spectrin beta non-erythrocytic 4 (SPTBN4) have been linked to congenital hypotonia, intellectual disability and motor neuropathy. Here we report on two siblings with a homozygous splice-site mutation in the SPTBN4 gene, lacking previously reported features of the disorder such as seizures, feeding difficulties, respiratory difficulties or profound intellectual disability. Our findings indicate that muscular hypotonia, myopathic facies with ptosis and axonal neuropathy can be the core clinical features in the SPTBN4 disorder and suggest that SPTBN4 mutation analysis should be considered in infants with marked axonal neuropathy.


Subject(s)
Axons/pathology , Hereditary Sensory and Motor Neuropathy/genetics , Muscle Hypotonia/genetics , Protein Isoforms/genetics , Spectrin/genetics , Axons/ultrastructure , Child , Child, Preschool , Female , Hereditary Sensory and Motor Neuropathy/pathology , Homozygote , Humans , Male , Mutation , Phenotype
16.
Sci Rep ; 9(1): 19336, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31852952

ABSTRACT

EGR2 (early growth response 2) is a crucial transcription factor for the myelination of the peripheral nervous system. Mutations in EGR2 are reported to cause a heterogenous spectrum of peripheral neuropathy with wide variation in both severity and age of onset, including demyelinating and axonal forms of Charcot-Marie Tooth (CMT) neuropathy, Dejerine-Sottas neuropathy (DSN/CMT3), and congenital hypomyelinating neuropathy (CHN/CMT4E). Here we report a sporadic de novo EGR2 variant, c.1232A > G (NM_000399.5), causing a missense p.Asp411Gly substitution and discovered through whole-exome sequencing (WES) of the proband. The resultant phenotype is severe demyelinating DSN with onset at two years of age, confirmed through nerve biopsy and electrophysiological examination. In silico analyses showed that the Asp411 residue is evolutionarily conserved, and the p.Asp411Gly variant was predicted to be deleterious by multiple in silico analyses. A luciferase-based reporter assay confirmed the reduced ability of p.Asp411Gly EGR2 to activate a PMP22 (peripheral myelin protein 22) enhancer element compared to wild-type EGR2. This study adds further support to the heterogeneity of EGR2-related peripheral neuropathies and provides strong functional evidence for the pathogenicity of the p.Asp411Gly EGR2 variant.


Subject(s)
Early Growth Response Protein 2/genetics , Genetic Predisposition to Disease , Hereditary Sensory and Motor Neuropathy/genetics , Mutation/genetics , Adolescent , Adult , Age of Onset , Amino Acid Sequence , Base Sequence , Child , Child, Preschool , Computer Simulation , Early Growth Response Protein 2/chemistry , Female , Hereditary Sensory and Motor Neuropathy/diagnostic imaging , Hereditary Sensory and Motor Neuropathy/pathology , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neural Conduction , Pedigree , Protein Domains , Schwann Cells/metabolism , Transcription, Genetic , Transcriptional Activation/genetics , Exome Sequencing
17.
Muscle Nerve ; 60(6): 739-744, 2019 12.
Article in English | MEDLINE | ID: mdl-31449671

ABSTRACT

BACKGROUND: Hereditary motor and sensory neuropathy with proximal dominant involvement (HMSN-P) is characterized by adult onset, a slowly progressive course and autosomal dominant inheritance. It remains unclear whether myopathic changes occur histopathologically. METHODS: We encountered 2 patients in a family with a heterozygous p.P285L mutation in TRK-fused gene (TFG), which is known to cause HMSN-P. The affected individuals developed proximal-dominant muscle weakness in their 40s, which slowly progressed to a motor neuron disease-like phenotype. RESULTS: Muscle biopsy showed myopathic pathology including fiber size variability, increased internal nuclei, fiber splitting, and core-like structures, associated with neurogenic changes: large groups of atrophic fibers and fiber type-grouping. Immunohistochemistry revealed sarcoplasmic aggregates of TFG, TDP-43, and p62 without congophilic material. CONCLUSIONS: The present study demonstrates myopathic changes in HMSN-P. Although the mechanisms underlying the skeletal muscle involvement remain to be elucidated, immunohistochemistry suggests that abnormal protein aggregation may be involved in the myopathic pathology.


Subject(s)
Hereditary Sensory and Motor Neuropathy/pathology , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Action Potentials , DNA-Binding Proteins/metabolism , Female , Fluorescent Antibody Technique , Hereditary Sensory and Motor Neuropathy/diagnostic imaging , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Muscle, Skeletal/diagnostic imaging , Muscular Atrophy/pathology , Neural Conduction , Pedigree , Proteins/genetics , RNA-Binding Proteins/metabolism , Sarcoplasmic Reticulum/metabolism , Siblings
18.
Ann Clin Transl Neurol ; 6(2): 401-405, 2019 02.
Article in English | MEDLINE | ID: mdl-30847374

ABSTRACT

Vaccinia-related kinase 1 (VRK1) mutations can cause motor phenotypes including axonal sensorimotor neuropathy, distal hereditary motor neuropathy (dHMN), spinal muscular atrophy, and amyotrophic lateral sclerosis. Here, we identify a novel homozygous VRK1 p.W375X mutation causing recessive dHMN. The proband presented with juvenile onset of weakness in the distal lower extremities, slowly progressing to the distal upper limbs, with bilateral pes cavus and no upper motor or sensory neuron involvement. Nerve conduction studies showed a pure motor axonal neuropathy. Our findings extend the ethnic distribution of VRK1 mutations, indicating that these mutations should be included in genetic diagnostic testing for dHMN.


Subject(s)
Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/pathology , Intracellular Signaling Peptides and Proteins/genetics , Muscular Atrophy, Spinal/genetics , Mutation/genetics , Protein Serine-Threonine Kinases/genetics , Adult , Hereditary Sensory and Motor Neuropathy/diagnosis , Humans , Male , Muscular Atrophy, Spinal/diagnosis , Pedigree , Phenotype
19.
Article in English | MEDLINE | ID: mdl-30889971

ABSTRACT

Objective: To describe a patient with facial onset sensory and motor neuronopathy (FOSMN) carrying heterozygous mutations in both TARDBP and SQSTM1 genes. Methods: The patient underwent neurological, neuropsychological, and neurophysiological examinations. Brain magnetic resonance imaging (MRI) and extensive genetic analysis were also performed. Results: The neurological examination showed dysphonia, left trigeminal hypesthesia, and left masseter and temporalis muscle atrophy. Mild cognitive impairment, affecting predominantly executive functions and social cognition, was appreciable in the neuropsychological examination. The electrophysiological studies revealed: left abnormal blink reflex; neurogenic changes in bulbar and cervical muscles; normal motor evoked potential amplitude, central motor conduction time and cortical silent period. Brain MRI showed right-predominant frontotemporal atrophy. Genetic analysis showed a heterozygous mutation in TARDBP (p.A390S) and in SQSTM1 (p.P392L), both previously described as causing amyotrophic lateral sclerosis. The SQSTM1, but not the TARDBP, mutation was found in both healthy siblings. Conclusions: Our data provide new clinical, neuroimaging, and genetic evidence that FOSMN is a neurodegenerative disease of the motor neuron disease and frontotemporal dementia spectrum, with a possible oligogenic origin. Multicentric efforts focusing on cognitive and genetic studies are necessary to confirm this hypothesis and to determine if ALS genes should be systematically screened in these patients.


Subject(s)
Facial Nerve Diseases/pathology , Hereditary Sensory and Motor Neuropathy/pathology , Motor Neuron Disease/pathology , Aged , Blinking , Cognitive Dysfunction/etiology , DNA Repeat Expansion , DNA-Binding Proteins/genetics , Electromyography , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/psychology , Humans , Magnetic Resonance Imaging , Male , Motor Neuron Disease/genetics , Motor Neuron Disease/psychology , Neurologic Examination , Neuropsychological Tests , Sequestosome-1 Protein/genetics , Tomography, X-Ray Computed
20.
Muscle Nerve ; 59(6): 665-670, 2019 06.
Article in English | MEDLINE | ID: mdl-30810227

ABSTRACT

INTRODUCTION: Onion-bulbs (OB) are concentrically layered Schwann-cell processes, surrounding nerve fibers, occurring in both inherited and acquired demyelinating polyneuropathies. We investigated whether OB patterns (generalized, mixed, or focal) correlate with acquired or inherited neuropathies. METHODS: One hundred thirty-one OB-rich nerve biopsies were graded for OB pattern and inflammation without knowledge of clinical history. We classified inherited (n = 49) or acquired (n = 82) neuropathies based solely on clinical history. RESULTS: Fifty-one biopsies had generalized (34 inherited vs. 17 acquired, P < 0.001), 54 mixed (48 acquired vs. 6 inherited, P < 0.001), and 26 focal/multifocal (inherited [n = 9], acquired [n = 17]) OB. Inflammation occurred more frequently in acquired (n = 54) than inherited (n = 14) neuropathy (P = 0.004). DISCUSSION: Generalized OB correlates with inherited neuropathy; mixed OB with acquired. Inflammation occurs more in acquired neuropathy cases. OB patterns are best explained by ubiquitous Schwann-cell involvement in inherited and multifocal Schwann-cell involvement in acquired neuropathies and predict the electrophysiology of uniform demyelination in inherited and unequal demyelination in acquired neuropathies. Muscle Nerve 59:665-670, 2019.


Subject(s)
Hereditary Sensory and Motor Neuropathy/pathology , Peripheral Nerves/pathology , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/pathology , Schwann Cells/pathology , Adolescent , Adult , Aged , Biopsy , Charcot-Marie-Tooth Disease/pathology , Female , Hereditary Sensory and Motor Neuropathy/genetics , Humans , Male , Middle Aged , Myelin Proteins/genetics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...