Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Diabetes Complications ; 37(2): 108383, 2023 02.
Article in English | MEDLINE | ID: mdl-36610321

ABSTRACT

Deoxysphingolipids (1-deoxySLs) are neurotoxic sphingolipids associated with obesity and diabetic neuropathy (DN) and have been linked to severity of functional peripheral neuropathies. While l-serine supplementation can reduce 1-deoxySL accumulation and improve insulin sensitivity and sensory nerve velocity, long-term outcomes have not yet been examined. To assess this, we treated 2 month old db/db mice, a model of DN, with 5-20 % oral l-serine for 6 months and longitudinally quantified the extent of functional neuropathy progression. We examined putative biomarkers of neuropathy in blood and tissue and quantified levels of small fiber neuropathy, looking for associations between lowered 1-deoxySL and phenotypes. Toxic 1-deoxySLs were suppressed long-term in plasma and various tissue including the sciatic nerve, which is particularly targeted in DN. Functional neuropathy and sensory modalities were significantly improved in the treatment group well into advanced stages of disease. However, structural assessments revealed prominent axonal degeneration, apoptosis and Schwann cell pathology, suggesting that neuropathy was ongoing. Hyperglycemia and dyslipidemia persisted during our study, and high levels of glutathione were seen in the spinal cord. Our results demonstrate that despite significant functional improvements, l-serine does not prevent chronic degenerative changes specifically at the structural level, pointing to other processes such as oxidative damage and hyperglycemia, that persist despite 1-deoxySL reduction.


Subject(s)
Diabetes Mellitus , Diabetic Neuropathies , Hyperglycemia , Mice , Animals , Serine/therapeutic use , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/prevention & control , Sphingolipids , Dietary Supplements
2.
Neuromolecular Med ; 21(2): 182-191, 2019 06.
Article in English | MEDLINE | ID: mdl-30955194

ABSTRACT

Hereditary sensory and autonomic neuropathy type 1 (HSAN1) is a rare, autosomal dominantly inherited, slowly progressive and length-dependent axonal peripheral neuropathy. HSAN1 is associated with several mutations in serine-palmitoyltransferase (SPT), the first enzyme in the de novo sphingolipid biosynthetic pathway. HSAN1 mutations alter the substrate specificity of SPT, which leads to the formation of 1-deoxysphingolipids, an atypical and neurotoxic subclass of sphingolipids. This study describes the clinical and neurophysiological phenotype of a German family with a novel SPTCL2 mutation (c.529A > G; N177D) associated with HSAN1 and the biochemical characterization of this mutation.) The mutaion was identified in five family members that segregated with the diesease. Patients were characterized genetically and clinically for neurophysiological function. Their plasma sphingolipid profiles were analyzed by LC-MS. The biochemical properties of the mutation were characterized in a cell-based activity assay. Affected family members showed elevated 1-deoxysphingolipid plasma levels. HEK293 cells expressing the N177D SPTLC2 mutant showed increased de novo 1-deoxysphingolipid formation, but also displayed elevated canonical SPT activity and increased C20 sphingoid base production. This study identifies the SPTLC2 N177D variant as a novel disease-causing mutation with increased 1-deoxySL formation and its association with a typical HSAN1 phenotype.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/genetics , Mutation, Missense , Point Mutation , Serine C-Palmitoyltransferase/genetics , Alanine/metabolism , Amino Acid Sequence , Consensus Sequence , Female , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Male , Models, Molecular , Pedigree , Protein Conformation , Retrospective Studies , Sequence Alignment , Sequence Homology, Amino Acid , Serine C-Palmitoyltransferase/deficiency , Serine C-Palmitoyltransferase/physiology , Sphingolipids/biosynthesis , Sphingolipids/blood
3.
J Neurol Neurosurg Psychiatry ; 90(8): 895-906, 2019 08.
Article in English | MEDLINE | ID: mdl-30995999

ABSTRACT

OBJECTIVES: Hereditary sensory neuropathy type 1 (HSN1) is a rare, slowly progressive neuropathy causing profound sensory deficits and often severe motor loss. L-serine supplementation is a possible candidate therapy but the lack of responsive outcome measures is a barrier for undertaking clinical trials in HSN1. We performed a 12-month natural history study to characterise the phenotype of HSN1 and to identify responsive outcome measures. METHODS: Assessments included Charcot-Marie-Tooth Neuropathy Score version 2 (CMTNSv2), CMTNSv2-Rasch modified, nerve conduction studies, quantitative sensory testing, intraepidermal nerve fibre density (thigh), computerised myometry (lower limbs), plasma 1-deoxysphingolipid levels, calf-level intramuscular fat accumulation by MRI and patient-based questionnaires (Neuropathic Pain Symptom Inventory and 36-Short Form Health Survey version 2 [SF-36v2]). RESULTS: 35 patients with HSN1 were recruited. There was marked heterogeneity in the phenotype mainly due to differences between the sexes: males generally more severely affected. The outcome measures that significantly changed over 1 year and correlated with CMTNSv2, SF-36v2-physical component and disease duration were MRI determined calf intramuscular fat accumulation (mean change in overall calf fat fraction 2.36%, 95% CI 1.16 to 3.55, p=0.0004), pressure pain threshold on the hand (mean change 40 kPa, 95% CI 0.7 to 80, p=0.046) and myometric measurements of ankle plantar flexion (median change -0.5 Nm, IQR -9.5 to 0, p=0.0007), ankle inversion (mean change -0.89 Nm, 95% CI -1.66 to -0.12, p=0.03) and eversion (mean change -1.61 Nm, 95% CI -2.72 to -0.51, p=0.006). Intramuscular calf fat fraction was the most responsive outcome measure. CONCLUSION: MRI determined calf muscle fat fraction shows validity and high responsiveness over 12 months and will be useful in HSN1 clinical trials.


Subject(s)
Adipose Tissue/diagnostic imaging , Hereditary Sensory and Autonomic Neuropathies , Magnetic Resonance Imaging , Muscle, Skeletal/diagnostic imaging , Outcome Assessment, Health Care , Predictive Value of Tests , Adult , Disease Progression , Female , Hereditary Sensory and Autonomic Neuropathies/diagnostic imaging , Hereditary Sensory and Autonomic Neuropathies/genetics , Humans , Lower Extremity/diagnostic imaging , Male , Phenotype , Surveys and Questionnaires
4.
J Clin Invest ; 129(3): 1229-1239, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30620338

ABSTRACT

BACKGROUND: Sphingolipids are important components of cellular membranes and functionally associated with fundamental processes such as cell differentiation, neuronal signaling, and myelin sheath formation. Defects in the synthesis or degradation of sphingolipids leads to various neurological pathologies; however, the entire spectrum of sphingolipid metabolism disorders remains elusive. METHODS: A combined approach of genomics and lipidomics was applied to identify and characterize a human sphingolipid metabolism disorder. RESULTS: By whole-exome sequencing in a patient with a multisystem neurological disorder of both the central and peripheral nervous systems, we identified a homozygous p.Ala280Val variant in DEGS1, which catalyzes the last step in the ceramide synthesis pathway. The blood sphingolipid profile in the patient showed a significant increase in dihydro sphingolipid species that was further recapitulated in patient-derived fibroblasts, in CRISPR/Cas9-derived DEGS1-knockout cells, and by pharmacological inhibition of DEGS1. The enzymatic activity in patient fibroblasts was reduced by 80% compared with wild-type cells, which was in line with a reduced expression of mutant DEGS1 protein. Moreover, an atypical and potentially neurotoxic sphingosine isomer was identified in patient plasma and in cells expressing mutant DEGS1. CONCLUSION: We report DEGS1 dysfunction as the cause of a sphingolipid disorder with hypomyelination and degeneration of both the central and peripheral nervous systems. TRIAL REGISTRATION: Not applicable. FUNDING: Seventh Framework Program of the European Commission, Swiss National Foundation, Rare Disease Initiative Zurich.


Subject(s)
Central Nervous System Diseases , Fatty Acid Desaturases , Lipid Metabolism, Inborn Errors , Mutation, Missense , Sphingosine , Amino Acid Substitution , Cell Line , Central Nervous System Diseases/enzymology , Central Nervous System Diseases/genetics , Central Nervous System Diseases/pathology , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/metabolism , Female , Humans , Lipid Metabolism, Inborn Errors/enzymology , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Male , Sphingosine/genetics , Sphingosine/metabolism , Exome Sequencing
5.
Neurology ; 92(4): e359-e370, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30626650

ABSTRACT

OBJECTIVE: To evaluate the safety and efficacy of l-serine in humans with hereditary sensory autonomic neuropathy type I (HSAN1). METHODS: In this randomized, placebo-controlled, parallel-group trial with open-label extension, patients aged 18-70 years with symptomatic HSAN1 were randomized to l-serine (400 mg/kg/day) or placebo for 1 year. All participants received l-serine during the second year. The primary outcome measure was the Charcot-Marie-Tooth Neuropathy Score version 2 (CMTNS). Secondary outcomes included plasma sphingolipid levels, epidermal nerve fiber density, electrophysiologic measurements, patient-reported measures, and adverse events. RESULTS: Between August 2013 and April 2014, we enrolled and randomized 18 participants, 16 of whom completed the study. After 1 year, the l-serine group experienced improvement in CMTNS relative to the placebo group (-1.5 units, 95% CI -2.8 to -0.1, p = 0.03), with evidence of continued improvement in the second year of treatment (-0.77, 95% CI -1.67 to 0.13, p = 0.09). Concomitantly, deoxysphinganine levels dropped in l-serine-treated but not placebo-treated participants (59% decrease vs 11% increase; p < 0.001). There were no serious adverse effects related to l-serine. CONCLUSION: High-dose oral l-serine supplementation appears safe in patients with HSAN1 and is potentially effective at slowing disease progression. CLINICALTRIALSGOV IDENTIFIER: NCT01733407. CLASSIFICATION OF EVIDENCE: This study provides Class I evidence that high-dose oral l-serine supplementation significantly slows disease progression in patients with HSAN1.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/drug therapy , Serine/therapeutic use , Treatment Outcome , Adolescent , Adult , Aged , Double-Blind Method , Female , Follow-Up Studies , Hereditary Sensory and Autonomic Neuropathies/etiology , Humans , Male , Middle Aged , Neural Conduction/drug effects , Pain Measurement , Serine C-Palmitoyltransferase/genetics , Sphingolipids/metabolism , Surveys and Questionnaires , Ubiquitin Thiolesterase/metabolism , Young Adult
6.
Article in English | MEDLINE | ID: mdl-29042446

ABSTRACT

Hereditary sensory neuropathy type 1 (HSAN1) may be the first genetic neuropathy amenable to a specific mechanism-based treatment, as L-serine supplementation can be used to lower the neurotoxic levels of 1-deoxysphingolipids (1-deoxySL) that cause the neurodegeneration. The treatment is so far untested in HSAN1C caused by variants in the serine palmitoyl transferase subunit 2 (SPTLC2) gene. The aim of this study was to establish whether oral L-serine lowers 1-deoxySL in a patient with HSAN1C, to perform a dose escalation to find the minimal effective dose, and to assess the safety profile and global metabolic effects of the treatment. Our patient underwent a 52-wk treatment in which the L-serine dose was titrated up to 400 mg/kg/day. She was followed up by repeated clinical examination, nerve conduction testing, and skin biopsies to document effects on small nerve fibers. Serum was assayed for 1-deoxySL and metabolomics analysis of 111 metabolites. We found a robust lowering of 1-deoxySL, which correlated in a near-linear fashion with increased serum L-serine levels. Metabolomics analysis showed a modest elevation in glycine and a marked reduction in the level of cytosine, whereas most of the other assayed metabolites did not change. There were no direct side effects from the treatment, but the patient developed a transitory toe ulceration during the course of the study. The Charcot-Marie-Tooth neuropathy score increased by 1 point. We conclude that oral supplementation of L-serine decreases 1-deoxySL in HSAN1C without major global effects on metabolism. L-serine is therefore a potential treatment for HSAN1C.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/diet therapy , Serine C-Palmitoyltransferase/genetics , Serine/therapeutic use , Adult , Dietary Supplements , Female , Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/metabolism , Humans , Mutation , Serine/metabolism , Serine C-Palmitoyltransferase/blood , Serine C-Palmitoyltransferase/metabolism , Sphingolipids/blood
7.
J Neurochem ; 143(5): 507-522, 2017 12.
Article in English | MEDLINE | ID: mdl-28902413

ABSTRACT

Hereditary neuropathies comprise a wide variety of chronic diseases associated to more than 80 genes identified to date. We herein examined 612 index patients with either a Charcot-Marie-Tooth phenotype, hereditary sensory neuropathy, familial amyloid neuropathy, or small fiber neuropathy using a customized multigene panel based on the next generation sequencing technique. In 121 cases (19.8%), we identified at least one putative pathogenic mutation. Of these, 54.4% showed an autosomal dominant, 33.9% an autosomal recessive, and 11.6% an X-linked inheritance. The most frequently affected genes were PMP22 (16.4%), GJB1 (10.7%), MPZ, and SH3TC2 (both 9.9%), and MFN2 (8.3%). We further detected likely or known pathogenic variants in HINT1, HSPB1, NEFL, PRX, IGHMBP2, NDRG1, TTR, EGR2, FIG4, GDAP1, LMNA, LRSAM1, POLG, TRPV4, AARS, BIC2, DHTKD1, FGD4, HK1, INF2, KIF5A, PDK3, REEP1, SBF1, SBF2, SCN9A, and SPTLC2 with a declining frequency. Thirty-four novel variants were considered likely pathogenic not having previously been described in association with any disorder in the literature. In one patient, two homozygous mutations in HK1 were detected in the multigene panel, but not by whole exome sequencing. A novel missense mutation in KIF5A was considered pathogenic because of the highly compatible phenotype. In one patient, the plasma sphingolipid profile could functionally prove the pathogenicity of a mutation in SPTLC2. One pathogenic mutation in MPZ was identified after being previously missed by Sanger sequencing. We conclude that panel based next generation sequencing is a useful, time- and cost-effective approach to assist clinicians in identifying the correct diagnosis and enable causative treatment considerations.


Subject(s)
Genetic Predisposition to Disease , Hereditary Sensory and Motor Neuropathy/genetics , Mutation/genetics , Rare Diseases/genetics , Charcot-Marie-Tooth Disease/genetics , Female , HSP27 Heat-Shock Proteins/genetics , Heat-Shock Proteins , Hereditary Sensory and Motor Neuropathy/diagnosis , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Molecular Chaperones , Phenotype
8.
J Tissue Eng Regen Med ; 11(2): 509-518, 2017 02.
Article in English | MEDLINE | ID: mdl-25066608

ABSTRACT

Aim of the study was to investigate the influence of the self-assembling peptide nanofibre scaffolds (SAPNs) on the growth, proliferation and retinal neuronal differentiation of the stem/progenitor cells (SCs) derived from the ciliary pigment epithelium (CPE) of human cadaveric eye. Here SAPNs (RADA16-I, PM), which is well described in previous studies, commercially available and xeno-free. The CPE cells isolated were cultured in DMEM/F12 supplemented with N2 and growth factors such as basic fibroblast growth factor and epidermal growth factor, encapsulated in the scaffolds. The entrapped SCs actively expanded and formed clone-like clusters in the scaffolds. Many cells in the cluster were proliferating, as revealed by 5-bromo-2-deoxyuridine uptake and could be maintained for up to 6 days and expressed neural progenitor markers such as ß-III tubulin, Nestin, Pax6 and Musashi1. Upon differentiation of these cells in conditioned medium, the cells exhibited retinal neuronal markers such as s-Opsin, rhodopsin and Recoverin. The RT2 profiler polymerase chain reaction array experiments showed selective gene expression, possibly involved in neural stem/progenitor cell adhesion and differentiation. These findings suggest the suitability of the three-dimensional culture system for the proliferation and maintenance of CPE stem/progenitor cells (CPE-NS) and for possible use in ex vivo studies of small molecules, drug deliveries for retinal diseases and for use in combination with directed stem/progenitor cell differentiation. and ultimately for tissue replacement therapies. Copyright © 2014 John Wiley & Sons, Ltd.


Subject(s)
Nanofibers/chemistry , Neural Stem Cells/cytology , Peptides/chemistry , Retinal Pigment Epithelium/cytology , Tissue Scaffolds/chemistry , Cadaver , Cell Adhesion , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Ciliary Body/cytology , Culture Media, Conditioned , Epithelial Cells/cytology , Fibroblast Growth Factor 2/metabolism , Humans , Retinal Neurons/metabolism
9.
Neuropharmacology ; 110(Pt A): 211-222, 2016 11.
Article in English | MEDLINE | ID: mdl-27016021

ABSTRACT

1-Deoxysphingolipids (1-deoxySL) are atypical and neurotoxic sphingolipids formed by alternate substrate usage of the enzyme serine-palmitoyltransferase. Pathologically increased 1-deoxySL formation causes hereditary sensory and autosomal neuropathy type 1 (HSAN1) - a progressive peripheral axonopathy. However, the underlying molecular mechanisms by which 1-deoxySL acts are unknown. Herein we studied the effect of 1-deoxysphinganine (1-deoxySA) and its canonical counterpart sphinganine (SA) in aged cultured neurons comparing their outcome on cell survival and cytoskeleton integrity. 1-deoxySA caused rapid neuronal cytoskeleton disruption and modulated important cytoskeletal regulatory and associated components including Rac1, Ezrin and insulin receptor substrate 53. We show that 1-deoxySA is internalized and metabolized downstream to 1-deoxydihydroceramide since inhibition of ceramide synthase protected neurons from 1-deoxySA-mediated cell death. In addition, 1-deoxySA reduced protein levels of N-methyl-d-aspartate receptor (NMDAR) subunit GluN2B, the postsynaptic density protein 95 and induced cleavage of p35 to p25. Notably, blocking NMDAR activation by MK-801 or memantine significantly prevented 1-deoxySA neurotoxicity. Functional studies of differentiating primary neurons via the patch-clamp technique demonstrated that 1-deoxySA irreversibly depolarizes the neuronal membrane potential in an age-dependent manner. Notably, only neuronal cells that displayed functional NMDAR- and NMDA-induced whole-cell currents responded to 1-deoxySA treatment. Furthermore, pre-exposure to the non-competitive antagonist MK-801 blocked the current response of NMDA and glycine, as well as 1-deoxySA. We conclude that 1-deoxySA-induced neurotoxicity compromises cytoskeletal stability and targets NMDAR signaling in an age-dependent manner. Thus stabilization of cytoskeletal structures and/or inhibition of glutamate receptors could be a potential therapeutic approach to prevent 1-deoxySA-induced neurodegeneration.


Subject(s)
Neurons/drug effects , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Sphingolipids/toxicity , Animals , Cell Line, Tumor , Cerebral Cortex , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Disks Large Homolog 4 Protein/metabolism , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Glycine/administration & dosage , Glycine/metabolism , Humans , Memantine/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice, Inbred C57BL , N-Methylaspartate/administration & dosage , N-Methylaspartate/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Sphingosine/toxicity , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/metabolism
10.
Hum Mol Genet ; 25(5): 853-65, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26681808

ABSTRACT

Hereditary sensory and autonomic neuropathy type 1 (HSAN1) is a rare autosomal dominant inherited peripheral neuropathy caused by mutations in the SPTLC1 and SPTLC2 subunits of serine palmitoyltransferase (SPT). The mutations induce a permanent shift in the substrate preference from L-serine to L-alanine, which results in the pathological formation of atypical and neurotoxic 1-deoxy-sphingolipids (1-deoxySL). Here we compared the enzymatic properties of 11 SPTLC1 and six SPTLC2 mutants using a uniform isotope labelling approach. In total, eight SPT mutants (STPLC1p.C133W, p.C133Y, p.S331F, p.S331Y and SPTLC2p.A182P, p.G382V, p.S384F, p.I504F) were associated with increased 1-deoxySL synthesis. Despite earlier reports, canonical activity with l-serine was not reduced in any of the investigated SPT mutants. Three variants (SPTLC1p.S331F/Y and SPTLC2p.I505Y) showed an increased canonical activity and increased formation of C20 sphingoid bases. These three mutations are associated with an exceptionally severe HSAN1 phenotype, and increased C20 sphingosine levels were also confirmed in plasma of patients. A principal component analysis of the analysed sphingoid bases clustered the mutations into three separate entities. Each cluster was related to a distinct clinical outcome (no, mild and severe HSAN1 phenotype). A homology model based on the protein structure of the prokaryotic SPT recapitulated the same grouping on a structural level. Mutations associated with the mild form clustered around the active site, whereas mutations associated with the severe form were located on the surface of the protein. In conclusion, we showed that HSAN1 mutations in SPT have distinct biochemical properties, which allowed for the prediction of the clinical symptoms on the basis of the plasma sphingoid base profile.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/genetics , Mutation , Serine C-Palmitoyltransferase/genetics , Adult , Aged , Alanine/chemistry , Alanine/metabolism , Catalytic Domain , Child , Gas Chromatography-Mass Spectrometry , Gene Expression , HEK293 Cells , Hereditary Sensory and Autonomic Neuropathies/enzymology , Hereditary Sensory and Autonomic Neuropathies/pathology , Humans , Isotope Labeling , Middle Aged , Models, Molecular , Phenotype , Principal Component Analysis , Recombinant Proteins/blood , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Serine/chemistry , Serine/metabolism , Serine C-Palmitoyltransferase/blood , Serine C-Palmitoyltransferase/chemistry , Severity of Illness Index , Sphingolipids/blood , Structural Homology, Protein , Structure-Activity Relationship , Young Adult
11.
Neuromolecular Med ; 18(1): 81-90, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26573920

ABSTRACT

Hereditary sensory and autonomic neuropathy 1 (HSAN1) is an autosomal dominant disorder that can be caused by variants in SPTLC1 or SPTLC2, encoding subunits of serine palmitoyl-CoA transferase. Disease variants alter the enzyme's substrate specificity and lead to accumulation of neurotoxic 1-deoxysphingolipids. We describe two families with autosomal dominant HSAN1C caused by a new variant in SPTLC2, c.547C>T, p.(Arg183Trp). The variant changed a conserved amino acid and was not found in public variant databases. All patients had a relatively mild progressive distal sensory impairment, with onset after age 50. Small fibers were affected early, leading to abnormalities on quantitative sensory testing. Sural biopsy revealed a severe chronic axonal neuropathy with subtotal loss of myelinated axons, relatively preserved number of non-myelinated fibers and no signs for regeneration. Skin biopsy with PGP9.5 labeling showed lack of intraepidermal nerve endings early in the disease. Motor manifestations developed later in the disease course, but there was no evidence of autonomic involvement. Patients had elevated serum 1-deoxysphingolipids, and the variant protein produced elevated amounts of 1-deoxysphingolipids in vitro, which proved the pathogenicity of the variant. Our results expand the genetic spectrum of HSAN1C and provide further detail about the clinical characteristics. Sequencing of SPTLC2 should be considered in all patients presenting with mild late-onset sensory-predominant small or large fiber neuropathy.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/genetics , Late Onset Disorders/genetics , Mutation, Missense , Serine C-Palmitoyltransferase/genetics , Age of Onset , Aged , Amino Acid Sequence , Amino Acid Substitution , Axons/pathology , Female , Finland , Genes, Dominant , Germany , Haplotypes , Humans , Male , Middle Aged , Molecular Sequence Data , Pedigree , Serine C-Palmitoyltransferase/deficiency , Serine C-Palmitoyltransferase/metabolism , Small Fiber Neuropathy/genetics , Sphingolipids/blood , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...