Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 895
Filter
Add more filters

Publication year range
1.
Immunity ; 50(5): 1218-1231.e5, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30952607

ABSTRACT

Patients with the neurological disorder HSAN-I suffer frequent infections, attributed to a lack of pain sensation and failure to seek care for minor injuries. Whether protective CD8+ T cells are affected in HSAN-I patients remains unknown. Here, we report that HSAN-I-associated mutations in serine palmitoyltransferase subunit SPTLC2 dampened human T cell responses. Antigen stimulation and inflammation induced SPTLC2 expression, and murine T-cell-specific ablation of Sptlc2 impaired antiviral-T-cell expansion and effector function. Sptlc2 deficiency reduced sphingolipid biosynthetic flux and led to prolonged activation of the mechanistic target of rapamycin complex 1 (mTORC1), endoplasmic reticulum (ER) stress, and CD8+ T cell death. Protective CD8+ T cell responses in HSAN-I patient PBMCs and Sptlc2-deficient mice were restored by supplementing with sphingolipids and pharmacologically inhibiting ER stress-induced cell death. Therefore, SPTLC2 underpins protective immunity by translating extracellular stimuli into intracellular anabolic signals and antagonizes ER stress to promote T cell metabolic fitness.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hereditary Sensory and Autonomic Neuropathies/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Mechanistic Target of Rapamycin Complex 1/metabolism , Serine C-Palmitoyltransferase/genetics , Animals , Cell Proliferation , Cells, Cultured , Cytokines/biosynthesis , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/immunology , Female , Humans , Lymphocytic Choriomeningitis/virology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Signal Transduction/immunology , Sphingolipids/biosynthesis
2.
Hum Mol Genet ; 32(8): 1380-1400, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36537577

ABSTRACT

A functional nerve growth factor NGF-Tropomyosin Receptor kinase A (TrkA) system is an essential requisite for the generation and maintenance of long-lasting thermal and mechanical hyperalgesia in adult mammals. Indeed, mutations in the gene encoding for TrkA are responsible for a rare condition, named Hereditary Sensory and Autonomic Neuropathy type IV (HSAN IV), characterized by the loss of response to noxious stimuli, anhidrosis and cognitive impairment. However, to date, there is no available mouse model to properly understand how the NGF-TrkA system can lead to pathological phenotypes that are distinctive of HSAN IV. Here, we report the generation of a knock-in mouse line carrying the HSAN IV TrkAR649W mutation. First, by in vitro biochemical and biophysical analyses, we show that the pathological R649W mutation leads to kinase-inactive TrkA also affecting its membrane dynamics and trafficking. In agreement with the HSAN IV human phenotype, TrkAR649W/m mice display a lower response to thermal and chemical noxious stimuli, correlating with reduced skin innervation, in addition to decreased sweating in comparison to TrkAh/m controls. Moreover, the R649W mutation decreases anxiety-like behavior and compromises cognitive abilities, by impairing spatial-working and social memory. Our results further uncover unexplored roles of TrkA in thermoregulation and sociability. In addition to accurately recapitulating the clinical manifestations of HSAN IV patients, our findings contribute to clarifying the involvement of the NGF-TrkA system in pain sensation.


Subject(s)
Disease Models, Animal , Hereditary Sensory and Autonomic Neuropathies , Receptor, trkA , Humans , Animals , Mice , Mutation , Receptor, trkA/genetics , Gene Knock-In Techniques , Nerve Growth Factor/metabolism , Phosphorylation , Genes, Lethal , Pain/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Skin/metabolism , Skin/pathology , Sympathetic Nervous System/metabolism , Hypohidrosis/metabolism , Behavior, Animal
3.
J Neurol Neurosurg Psychiatry ; 95(2): 103-113, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38041679

ABSTRACT

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease of the upper and lower motor neurons with varying ages of onset, progression and pathomechanisms. Monogenic childhood-onset ALS, although rare, forms an important subgroup of ALS. We recently reported specific SPTLC1 variants resulting in sphingolipid overproduction as a cause for juvenile ALS. Here, we report six patients from six independent families with a recurrent, de novo, heterozygous variant in SPTLC2 c.778G>A [p.Glu260Lys] manifesting with juvenile ALS. METHODS: Clinical examination of the patients along with ancillary and genetic testing, followed by biochemical investigation of patients' blood and fibroblasts, was performed. RESULTS: All patients presented with early-childhood-onset progressive weakness, with signs and symptoms of upper and lower motor neuron degeneration in multiple myotomes, without sensory neuropathy. These findings were supported on ancillary testing including nerve conduction studies and electromyography, muscle biopsies and muscle ultrasound studies. Biochemical investigations in plasma and fibroblasts showed elevated levels of ceramides and unrestrained de novo sphingolipid synthesis. Our studies indicate that SPTLC2 variant [c.778G>A, p.Glu260Lys] acts distinctly from hereditary sensory and autonomic neuropathy (HSAN)-causing SPTLC2 variants by causing excess canonical sphingolipid biosynthesis, similar to the recently reported SPTLC1 ALS associated pathogenic variants. Our studies also indicate that serine supplementation, which is a therapeutic in SPTLC1 and SPTCL2-associated HSAN, is expected to exacerbate the excess sphingolipid synthesis in serine palmitoyltransferase (SPT)-associated ALS. CONCLUSIONS: SPTLC2 is the second SPT-associated gene that underlies monogenic, juvenile ALS and further establishes alterations of sphingolipid metabolism in motor neuron disease pathogenesis. Our findings also have important therapeutic implications: serine supplementation must be avoided in SPT-associated ALS, as it is expected to drive pathogenesis further.


Subject(s)
Amyotrophic Lateral Sclerosis , Hereditary Sensory and Autonomic Neuropathies , Neurodegenerative Diseases , Child , Humans , Amyotrophic Lateral Sclerosis/genetics , Sphingolipids , Serine C-Palmitoyltransferase/genetics , Serine C-Palmitoyltransferase/metabolism , Hereditary Sensory and Autonomic Neuropathies/genetics , Serine
4.
Am J Med Genet A ; 194(7): e63579, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38436550

ABSTRACT

Due to the majority of currently available genome data deriving from individuals of European ancestry, the clinical interpretation of genomic variants in individuals from diverse ethnic backgrounds remains a major diagnostic challenge. Here, we investigated the genetic cause of a complex neurodevelopmental phenotype in two Palestinian siblings. Whole exome sequencing identified a homozygous missense TECPR2 variant (Chr14(GRCh38):g.102425085G>A; NM_014844.5:c.745G>A, p.(Gly249Arg)) absent in gnomAD, segregating appropriately with the inheritance pattern in the family. Variant assessment with in silico pathogenicity prediction and protein modeling tools alongside population database frequencies led to classification as a variant of uncertain significance. As pathogenic TECPR2 variants are associated with hereditary sensory and autonomic neuropathy with intellectual disability, we reviewed previously published candidate TECPR2 missense variants to clarify clinical outcomes and variant classification using current approved guidelines, classifying a number of published variants as of uncertain significance. This work highlights genomic healthcare inequalities and the challenges in interpreting rare genetic variants in populations underrepresented in genomic databases. It also improves understanding of the clinical and genetic spectrum of TECPR2-related neuropathy and contributes to addressing genomic data disparity and inequalities of the genomic architecture in Palestinian populations.


Subject(s)
Exome Sequencing , Hereditary Sensory and Autonomic Neuropathies , Mutation, Missense , Pedigree , Siblings , Humans , Male , Female , Mutation, Missense/genetics , Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/pathology , Hereditary Sensory and Autonomic Neuropathies/diagnosis , Phenotype , Child , Arabs/genetics , Genetic Predisposition to Disease , Homozygote
5.
Brain ; 146(12): 4880-4890, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37769650

ABSTRACT

Congenital insensitivity to pain (CIP) and hereditary sensory and autonomic neuropathies (HSAN) are clinically and genetically heterogeneous disorders exclusively or predominantly affecting the sensory and autonomic neurons. Due to the rarity of the diseases and findings based mainly on single case reports or small case series, knowledge about these disorders is limited. Here, we describe the molecular workup of a large international cohort of CIP/HSAN patients including patients from normally under-represented countries. We identify 80 previously unreported pathogenic or likely pathogenic variants in a total of 73 families in the >20 known CIP/HSAN-associated genes. The data expand the spectrum of disease-relevant alterations in CIP/HSAN, including novel variants in previously rarely recognized entities such as ATL3-, FLVCR1- and NGF-associated neuropathies and previously under-recognized mutation types such as larger deletions. In silico predictions, heterologous expression studies, segregation analyses and metabolic tests helped to overcome limitations of current variant classification schemes that often fail to categorize a variant as disease-related or benign. The study sheds light on the genetic causes and disease-relevant changes within individual genes in CIP/HSAN. This is becoming increasingly important with emerging clinical trials investigating subtype or gene-specific treatment strategies.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies , Pain Insensitivity, Congenital , Humans , Pain Insensitivity, Congenital/genetics , Hereditary Sensory and Autonomic Neuropathies/genetics , Mutation/genetics
6.
Pediatr Dermatol ; 41(1): 80-83, 2024.
Article in English | MEDLINE | ID: mdl-37345838

ABSTRACT

Congenital insensitivity to pain (CIP) is a rare phenotype characterized by the inability to perceive pain stimuli with subsequent self-injuries, whereas CIP associated with anhidrosis (CIPA) is an overlapping phenotype mainly characterized by insensitivity to noxious stimuli and anhidrosis. CIP is primarily associated with pathogenetic variants in the SCN9A gene while CIPA is associated with pathogenetic variants in NGF and NRTK genes. However, in recent years, a significant overlap between these two disorders has been observed highlighting the presence of anhidrosis in SCN9A variants. We report the cases of two siblings (age 4 and 6 years) born from consanguineous parents presenting with a previously undescribed phenotype due to a novel pathogenic variant in SCN9A clinically characterized by congenital insensitivity to pain, anhidrosis, and mild cognitive impairment.


Subject(s)
Channelopathies , Cognitive Dysfunction , Hereditary Sensory and Autonomic Neuropathies , Hypohidrosis , Indoles , Pain Insensitivity, Congenital , Propionates , Humans , Child, Preschool , Child , Pain Insensitivity, Congenital/genetics , Hypohidrosis/genetics , Mutation , Receptor, trkA/genetics , Pain/genetics , Cognitive Dysfunction/genetics , Hereditary Sensory and Autonomic Neuropathies/genetics , NAV1.7 Voltage-Gated Sodium Channel/genetics
7.
Ann Hum Genet ; 87(4): 147-157, 2023 07.
Article in English | MEDLINE | ID: mdl-36856139

ABSTRACT

Introduction Hereditary sensory neuropathy (HSN) describes as a heterogeneous group of peripheral neuropathies. HSN type 1 (HSN1) is one subtype characterized by distal sensory impairment that occurs in the form of numbness, tingling, or pain. To date, only two variants in the atlastin GTPase 3 (ATL3) gene have been identified that result in hereditary sensory neuropathy type 1F (HSN1F) with autosomal dominantinheritance. Methods We sudied and examined who present with sensory disturbances and muscle weakness in their lower limb. Patients underwent Whole Exome Sequencing and Sanger sequencing was performed in families for validation of detected variant. Results Here, we identified two Iranian families carrying the novel heterozygous stop variant NM_015459.5: c.16C>T, p.Arg6Ter in ATL3 that led to disturbed pain and touch sensitivity. This variant in the ATL3 gene was detected in both families (NM_015459.5: c.16C>T, p.Arg6Ter) by whole-exome sequencing and confirmed by Sanger sequencing. Conclusion In this study, the subjects manifested weakness of distal limb muscles and numbness of the lower extremities. In addition, some unusual features, including hearing problems and inability to sit and walk presented in one of the patients. Eventually, we provide a case-based review of the clinical features associated with HSN1F. Hitherto, only 11 patients with HSN1F have been reported. We compared our findings to previously reported cases, suggesting that the clinical features are generally variable in the HSN1F patients.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies , Peripheral Nervous System Diseases , Humans , Hypesthesia/genetics , Iran , Muscle Weakness/genetics , Pain/genetics , Pedigree , GTP Phosphohydrolases/genetics
8.
Clin Genet ; 104(5): 587-592, 2023 11.
Article in English | MEDLINE | ID: mdl-37431644

ABSTRACT

Arthrogryposis multiplex congenita (AMC) is a developmental condition characterized by multiple joint contractures resulting from reduced or absent fetal movements. Through whole-exome sequencing combined with arrayCGH from DNA of a fetus presenting with early onset AMC, we identified biallelic loss of function variants in Dystonin (DST): a stop gain variant (NM_001144769.5:c.12208G > T:p.(Glu4070Ter)) on the neuronal isoform and a 175 kb microdeletion including exons 25-96 of this isoform on the other allele [NC_000006.11:g.(56212278_56323554)_(56499398_56507586)del]. Transmission electron microscopy of the sciatic nerve revealed abnormal morphology of the peripheral nerve with severe hypomyelination associated with dramatic reduction of fiber density which highlights the critical role of DST in peripheral nerve axonogenesis during development in human. Variants in the neuronal isoforms of DST cause hereditary sensory and autonomic neuropathy which has been reported in several unrelated families with highly variable age of onset from fetal to adult onset. Our data enlarge the disease mechanisms of neurogenic AMC.


Subject(s)
Arthrogryposis , Hereditary Sensory and Autonomic Neuropathies , Adult , Humans , Pregnancy , Female , Arthrogryposis/diagnosis , Arthrogryposis/genetics , Dystonin/genetics , Protein Isoforms
9.
J Peripher Nerv Syst ; 28(3): 351-358, 2023 09.
Article in English | MEDLINE | ID: mdl-37448294

ABSTRACT

BACKGROUND AND AIMS: Homozygous loss-of-function mutations in the RETREG1 gene result in Hereditary Sensory Autonomic Neuropathy Type 2B. Clinical features include pain loss, autonomic disturbances, and upper motor neuron features. METHODS: We evaluated the clinical and genetic features of seven patients from four families with RETREG1 variants. RESULTS: Five patients were male. The median age of disease onset was 7.00 ± 2.81 (between 2 and 10 years). A combination of painless wounds, trophic changes, and foot ulcerations was the presenting symptom in five patients and walking difficulties in two. Motor symptoms were present in five patients. In a median disease duration of 30.00 ± 12.88 years, five patients had osteomyelitis, and three had toe amputations. A history of renal disease was present in one family. In another family, three affected siblings had short stature and a history of delayed puberty. Although sensory signs predominated the clinical findings, various degrees of motor signs such as muscle weakness, spasticity, and brisk tendon reflexes were noted in all patients. Nerve conduction studies showed axonal sensory-motor neuropathy in five patients and sensory neuropathy in two. Three pathogenic variants were identified in the RETREG1 gene. Two unrelated patients had a homozygous c.433C > T/p.(Gln145*), one a homozygous c.826delA/p.(Ser276Valfs*8), and the last had a novel homozygous c.102delC/p.(Ala35Glnfs*349) variants. INTERPRETATION: In our study, all patients showed signs and symptoms consistent with pain insensitivity. Although shadowed by sensory symptoms, motor signs were noted in our patients. Further studies are necessary to clarify the causal relationship between extra-neurological features and RETREG1 mutations.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies , Humans , Male , Child, Preschool , Child , Female , Pain , Muscle Weakness
10.
Mol Biol Rep ; 50(7): 6005-6017, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37273064

ABSTRACT

BACKGROUND: Family with sequence similarity 134, member B (FAM134B), also known as Reticulophagy regulator 1 (RETREG1), is an ER-phagy receptor involved in ER homeostasis. Congenital mutations in the FAM134B gene have been reported to be associated with hereditary sensory and autonomic neuropathy type 2B (HSAN2B); however, the molecular differences between wild-type and HSAN2B-linked FAM134B are not fully understood. METHODS AND RESULTS: We prepared several human FAM134B constructs, such as the HSAN2B-linked mutant, and compared their features with those of wild-type FAM134B by transfecting these constructs into FAM134B-deficient Neuro2a cells. Although intrinsic FAM134B protein expression in wild-type Neuro2a cells was affected by the supply of amino acids in the culture medium, the expression of each HSAN2B-linked mutant FAM134B protein was hardly affected by serum and amino acid deprivation. On the other hand, the intracellular localization of GFP-tagged HSAN2B-linked mutants, except for P7Gfs133X, overlapped well with ER-localized SP-RFPKDEL and did not differ from that of GFP-tagged wild-type FAM134B. However, analysis of protein‒protein interactions using the NanoBiT reporter assay revealed the difference between wild-type and C-terminal truncated mutant FAM134B. Furthermore, this NanoBiT assay demonstrated that both wild-type and G216R FAM134B interacted with LC3/GABARAPL1 to the same extent, but the FAM134B construct with mutations near the LC3-interacting region (LIR) did not. Similar to the NanoBiT assay, the C-terminal-truncated FAM134B showed lower ER-phagy activities, as assessed by the cotransfection of GFP-tagged reporters. CONCLUSIONS: We showed that wild-type and HSAN2B-linked FAM134B have different molecular characteristics by transfecting cells with various types of constructs. Thus, this study provides new insights into the molecular mechanisms underlying HSAN2B as well as the regulation of ER-phagy.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies , Intracellular Signaling Peptides and Proteins , Humans , Autophagy/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism
11.
Neurol Sci ; 44(7): 2551-2554, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36964315

ABSTRACT

We report a patient with early-onset hereditary sensory and autonomic neuropathy type 1A (HSAN-1A) who developed a distinct phenotype, with tongue fasciculation and atrophy, due to a mutation at serine 331 in the SPTLC1 gene. HSAN-1A manifestation causing tongue fasciculation and atrophy have been rarely found. Our report adds to the growing evidence of the existence of an overlap between hereditary neuropathy and motor neuron disease caused by pathogenic p.S331Y variant in SPTLC1 gene.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies , Motor Neuron Disease , Humans , Serine C-Palmitoyltransferase/genetics , Fasciculation , Phenotype , Hereditary Sensory and Autonomic Neuropathies/diagnosis , Hereditary Sensory and Autonomic Neuropathies/genetics , Mutation/genetics , Motor Neuron Disease/complications , Motor Neuron Disease/genetics , Atrophy
12.
Neurol Sci ; 44(4): 1415-1418, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36648562

ABSTRACT

INTRODUCTION: NAGLU encodes N-acetyl-alpha-glucosaminidase, an enzyme that degrades heparan sulfate. Biallelic NAGLU mutations cause mucopolysaccharidosis IIIB, a severe childhood-onset neurodegenerative disease, while monoallelic mutations are associated to late-onset, dominantly inherited painful sensory neuropathy. However, to date, only one family with a dominant NAGLU-related neuropathy has been described. CASE REPORT: Here we describe a patient with early-onset motor polyneuropathy harboring a novel monoallelic NAGLU mutation. We found reduced NAGLU enzymatic activity thus corroborating the pathogenic role of the new variant. DISCUSSION: Our report represents the second ever described case with dominant NAGLU-related neuropathy and the first case with early-onset motor symptoms. We underlie the importance of a thorough clinical description of this probably underestimated new clinical entity.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies , Mucopolysaccharidosis III , Neurodegenerative Diseases , Peripheral Nervous System Diseases , Polyneuropathies , Humans , Child , Mucopolysaccharidosis III/diagnosis , Mucopolysaccharidosis III/genetics , Mucopolysaccharidosis III/pathology , Polyneuropathies/genetics , Mutation/genetics
13.
Pediatr Int ; 65(1): e15415, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36371639

ABSTRACT

BACKGROUND: Novel coronavirus disease (COVID-19) outbreaks have dramatically changed lifestyles, with various effects on the physical and mental health of families and children with various childhood-onset neurological diseases. A questionnaire survey was conducted to identify family-specific issues and needs of patients with congenital insensitivity to pain with anhidrosis (CIPA) during major changes in their daily lives due to the COVID-19 outbreaks. METHODS: An anonymous questionnaire was sent to 56 families that were members of the Association of Patients and Families of CIPA in Japan between October and November 2020, the first 2 months of the third outbreak. RESULTS: Thirty-eight families (67.2% response rate) responded to the questionnaire. The current concerns of the parents were (1) difficulty in predicting the future (19 parents, 50%), (2) household and work concerns (eight parents, 21.1%), and (3) whether they would become infected (25 parents, 65.8%). Fifteen families indicated stress due to increased time together (stress + group), and 10 families had a better understanding of each other due to increased time together. New sleep disturbances and behavioral changes were observed in approximately 20% and 50% of patients with CIPA, respectively. No single factor could explain family stress. There were also free descriptions of the importance of peer support, connections with experts, and prompt responses for resolution. CONCLUSIONS: Each family has its own way of coping with multiple factors that contribute to the stress of the patient and family. A long-established resilience to the disease proved effective during this pandemic.


Subject(s)
COVID-19 , Hereditary Sensory and Autonomic Neuropathies , Child , Humans , Pandemics , Receptor, trkA , COVID-19/epidemiology
14.
Hum Mol Genet ; 29(11): 1864-1881, 2020 07 21.
Article in English | MEDLINE | ID: mdl-31984424

ABSTRACT

ADCA-DN and HSN-IE are rare neurodegenerative syndromes caused by dominant mutations in the replication foci targeting sequence (RFTS) of the DNA methyltransferase 1 (DNMT1) gene. Both phenotypes resemble mitochondrial disorders, and mitochondrial dysfunction was first observed in ADCA-DN. To explore mitochondrial involvement, we studied the effects of DNMT1 mutations in fibroblasts from four ADCA-DN and two HSN-IE patients. We documented impaired activity of purified DNMT1 mutant proteins, which in fibroblasts results in increased DNMT1 amount. We demonstrated that DNMT1 is not localized within mitochondria, but it is associated with the mitochondrial outer membrane. Concordantly, mitochondrial DNA failed to show meaningful CpG methylation. Strikingly, we found activated mitobiogenesis and OXPHOS with significant increase of H2O2, sharply contrasting with a reduced ATP content. Metabolomics profiling of mutant cells highlighted purine, arginine/urea cycle and glutamate metabolisms as the most consistently altered pathways, similar to primary mitochondrial diseases. The most severe mutations showed activation of energy shortage AMPK-dependent sensing, leading to mTORC1 inhibition. We propose that DNMT1 RFTS mutations deregulate metabolism lowering ATP levels, as a result of increased purine catabolism and urea cycle pathways. This is associated with a paradoxical mitochondrial hyper-function and increased oxidative stress, possibly resulting in neurodegeneration in non-dividing cells.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/genetics , Genetic Predisposition to Disease , Hereditary Sensory and Autonomic Neuropathies/genetics , Nerve Degeneration/genetics , Spinocerebellar Ataxias/genetics , DNA Methylation/genetics , Deafness/genetics , Deafness/physiopathology , Female , Fibroblasts/metabolism , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Humans , Male , Mitochondria/genetics , Mitochondria/metabolism , Mutation/genetics , Narcolepsy/genetics , Narcolepsy/physiopathology , Nerve Degeneration/physiopathology , Oxidative Phosphorylation , Phenotype , Protein Processing, Post-Translational/genetics , Spinocerebellar Ataxias/physiopathology
15.
N Engl J Med ; 381(15): 1422-1433, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31509666

ABSTRACT

BACKGROUND: Identifying mechanisms of diseases with complex inheritance patterns, such as macular telangiectasia type 2, is challenging. A link between macular telangiectasia type 2 and altered serine metabolism has been established previously. METHODS: Through exome sequence analysis of a patient with macular telangiectasia type 2 and his family members, we identified a variant in SPTLC1 encoding a subunit of serine palmitoyltransferase (SPT). Because mutations affecting SPT are known to cause hereditary sensory and autonomic neuropathy type 1 (HSAN1), we examined 10 additional persons with HSAN1 for ophthalmologic disease. We assayed serum amino acid and sphingoid base levels, including levels of deoxysphingolipids, in patients who had macular telangiectasia type 2 but did not have HSAN1 or pathogenic variants affecting SPT. We characterized mice with low serine levels and tested the effects of deoxysphingolipids on human retinal organoids. RESULTS: Two variants known to cause HSAN1 were identified as causal for macular telangiectasia type 2: of 11 patients with HSAN1, 9 also had macular telangiectasia type 2. Circulating deoxysphingolipid levels were 84.2% higher among 125 patients with macular telangiectasia type 2 who did not have pathogenic variants affecting SPT than among 94 unaffected controls. Deoxysphingolipid levels were negatively correlated with serine levels, which were 20.6% lower than among controls. Reduction of serine levels in mice led to increases in levels of retinal deoxysphingolipids and compromised visual function. Deoxysphingolipids caused photoreceptor-cell death in retinal organoids, but not in the presence of regulators of lipid metabolism. CONCLUSIONS: Elevated levels of atypical deoxysphingolipids, caused by variant SPTLC1 or SPTLC2 or by low serine levels, were risk factors for macular telangiectasia type 2, as well as for peripheral neuropathy. (Funded by the Lowy Medical Research Institute and others.).


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/genetics , Mutation , Retinal Telangiectasis/genetics , Serine C-Palmitoyltransferase/genetics , Serine/metabolism , Sphingolipids/metabolism , Adult , Aged , Animals , DNA Mutational Analysis , Disease Models, Animal , Exome/genetics , Female , Hereditary Sensory and Autonomic Neuropathies/complications , Hereditary Sensory and Autonomic Neuropathies/metabolism , Humans , Lipid Metabolism , Macula Lutea/pathology , Male , Mice , Middle Aged , Pedigree , Retinal Telangiectasis/complications , Retinal Telangiectasis/metabolism , Risk Factors , Serine/blood , Sphingosine/analogs & derivatives , Sphingosine/analysis , Young Adult
16.
Neuropathol Appl Neurobiol ; 48(7): e12842, 2022 12.
Article in English | MEDLINE | ID: mdl-35904184

ABSTRACT

AIMS: SPTLC1-related disorder is a late onset sensory-autonomic neuropathy associated with perturbed sphingolipid homeostasis which can be improved by supplementation with the serine palmitoyl-CoA transferase (SPT) substrate, l-serine. Recently, a juvenile form of motor neuron disease has been linked to SPTLC1 variants. Variants affecting the p.S331 residue of SPTLC1 cause a distinct phenotype, whose pathogenic basis has not been established. This study aims to define the neuropathological and biochemical consequences of the SPTLC1 p.S331 variant, and test response to l-serine in this specific genotype. METHODS: We report clinical and neurophysiological characterisation of two unrelated children carrying distinct p.S331 SPTLC1 variants. The neuropathology was investigated by analysis of sural nerve and skin innervation. To clarify the biochemical consequences of the p.S331 variant, we performed sphingolipidomic profiling of serum and skin fibroblasts. We also tested the effect of l-serine supplementation in skin fibroblasts of patients with p.S331 mutations. RESULTS: In both patients, we recognised an early onset phenotype with prevalent progressive motor neuron disease. Neuropathology showed severe damage to the sensory and autonomic systems. Sphingolipidomic analysis showed the coexistence of neurotoxic deoxy-sphingolipids with an excess of canonical products of the SPT enzyme. l-serine supplementation in patient fibroblasts reduced production of toxic 1-deoxysphingolipids but further increased the overproduction of sphingolipids. CONCLUSIONS: Our findings suggest that p.S331 SPTLC1 variants lead to an overlap phenotype combining features of sensory and motor neuropathies, thus proposing a continuum in the spectrum of SPTLC1-related disorders. l-serine supplementation in these patients may be detrimental.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies , Motor Neuron Disease , Peripheral Nervous System Diseases , Humans , Serine C-Palmitoyltransferase/chemistry , Serine C-Palmitoyltransferase/genetics , Mutation , Sphingolipids , Serine/chemistry , Serine/genetics
17.
J Anat ; 241(5): 1169-1185, 2022 Nov.
Article in English | MEDLINE | ID: mdl-34875719

ABSTRACT

Animal models of neurodegenerative diseases such as inherited peripheral neuropathies sometimes accurately recreate the pathophysiology of the human disease, and sometimes accurately recreate the genetic perturbations found in patients. Ideally, models achieve both, but this is not always possible; nonetheless, such models are informative. Here we describe two animal models of inherited peripheral neuropathy: mice with a mutation in tyrosyl tRNA-synthetase, YarsE196K , modeling dominant intermediate Charcot-Marie-Tooth disease type C (diCMTC), and mice with a mutation in serine palmitoyltransferase long chain 1, Sptlc1C133W , modeling hereditary sensory and autonomic neuropathy type 1 (HSAN1). YarsE196K mice develop disease-relevant phenotypes including reduced motor performance and reduced nerve conduction velocities by 4 months of age. Peripheral motor axons are reduced in size, but there is no reduction in axon number and plasma neurofilament light chain levels are not increased. Unlike the dominant human mutations, the YarsE196K mice only show these phenotypes as homozygotes, or as compound heterozygotes with a null allele, and no phenotype is observed in E196K or null heterozygotes. The Sptlc1C133W mice carry a knockin allele and show the anticipated increase in 1-deoxysphingolipids in circulation and in a variety of tissues. They also have mild behavioral defects consistent with HSAN1, but do not show neurophysiological defects or axon loss in peripheral nerves or in the epidermis of the hind paw or tail. Thus, despite the biochemical phenotype, the Sptlc1C133W mice do not show a strong neuropathy phenotype. Surprisingly, these mice were lethal as homozygotes, but the heterozygous genotype studied corresponds to the dominant genetics seen in humans. Thus, YarsE196K homozygous mice have a relevant phenotype, but imprecisely reproduce the human genetics, whereas the Sptlc1C133W mice precisely reproduce the human genetics, but do not recreate the disease phenotype. Despite these shortcomings, both models are informative and will be useful for future research.


Subject(s)
Charcot-Marie-Tooth Disease , Hereditary Sensory and Autonomic Neuropathies , Animals , Charcot-Marie-Tooth Disease/genetics , Disease Models, Animal , Hereditary Sensory and Autonomic Neuropathies/genetics , Humans , Ligases/genetics , Mice , Mutation , Peripheral Nervous System Diseases , RNA, Transfer , Serine C-Palmitoyltransferase/genetics
18.
Exp Dermatol ; 31(6): 949-955, 2022 06.
Article in English | MEDLINE | ID: mdl-35276021

ABSTRACT

DST encodes bullous pemphigoid antigen-1 (BPAG1), a protein with eight tissue-specific isoforms expressed in the skin, muscle, brain and nerves. Accordingly, mutations in this gene are associated with epidermolysis bullosa simplex (EBS) and hereditary sensory and autonomic neuropathy type 6 (HSAN-VI). The genotypic spectrum is attested to by 19 distinct mutations but genotype-phenotype correlation for both disorders is not well established. In this study, we performed next-generation sequencing (NGS) on two families with different phenotypic presentations, one foetus (P1) with musculoskeletal and neurological malformations established by prenatal ultrasound and family history, and a 15-year-old female patient (P2) with skin blistering. P1 had a novel homozygous nonsense mutation, DST: NM_001144769, c.3805C>T, p.R1269* within a region of genetic homozygosity (ROH). This mutation resides within the plakin domain of BPAG1 and ablates all isoforms of this protein, leading to novel extracutaneous phenotypes consistent with HSAN-VI in P1. P2 had a recurrent homozygous mutation DST: NM_001723.7, c.3370C>T, p.Gln1124* that presented with giant, trauma-induced skin blisters without extracutaneous involvement. This mutation is located within the coiled-coil domain present in the skin isoform of DST, BPGA1-e, associated with EBS. In summary, we report two families with pathogenic DST variants and expand the spectrum of DST genotype and phenotypes.


Subject(s)
Dystonin , Epidermolysis Bullosa Simplex , Hereditary Sensory and Autonomic Neuropathies , Dystonin/genetics , Epidermolysis Bullosa Simplex/genetics , Epidermolysis Bullosa Simplex/metabolism , Female , Hereditary Sensory and Autonomic Neuropathies/genetics , Homozygote , Humans , Mutation , Phenotype , Protein Isoforms/genetics
19.
Am J Med Genet A ; 188(4): 1245-1250, 2022 04.
Article in English | MEDLINE | ID: mdl-34897952

ABSTRACT

The DST gene is located on chromosome 6p and encodes for a large protein. Alternative splicing of this protein produces the neuronal (a1-a3), muscular (b1-b3), and epithelial (e) isoforms. Hereditary sensory and autonomic neuropathy (HSAN) type VI is a rare autosomal recessive disorder due to mutations affecting the a2 isoform. We present a case of HSAN-VI in a male neonate born to consanguineous parents. Genome sequencing revealed a novel homozygous variant (DST_c.1118C > T; p.Pro373Leu) inherited from both parents. This case further expands the phenotype and genotype of this rare syndrome.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies , Dystonin/genetics , Hereditary Sensory and Autonomic Neuropathies/diagnosis , Hereditary Sensory and Autonomic Neuropathies/genetics , Humans , Infant , Male , Neurons/metabolism , Phenotype , Protein Isoforms/genetics
20.
Am J Med Genet A ; 188(7): 2153-2161, 2022 07.
Article in English | MEDLINE | ID: mdl-35332675

ABSTRACT

Hereditary sensory and autonomic neuropathy type 2B (HSAN2B) is a rare autosomal recessive peripheral neuropathy caused by biallelic variants in RETREG1 (formerly FAM134B). HSAN2B is characterized by sensory impairment resulting in skin ulcerations, amputations, and osteomyelitis as well as variable weakness, spasticity, and autonomic dysfunction. Here, we report four affected individuals with recurrent osteomyelitis, ulceration, and amputation of hands and feet, sensory neuropathy, hyperhidrosis, urinary incontinence, and renal failure from a family without any known shared parental ancestry. Due to the history of chronic recurrent multifocal osteomyelitis and microcytic anemia, a diagnosis of Majeed syndrome was considered; however, sequencing of LPIN2 was negative. Family-based exome sequencing (ES) revealed a novel homozygous ultrarare RETREG1 variant NM_001034850.2:c.321G>A;p.Trp107Ter. Electrophysiological studies of the proband demonstrated axonal sensorimotor neuropathy predominantly in the lower extremities. Consistent with the lack of shared ancestry, the coefficient of inbreeding calculated from ES data was low (F = 0.002), but absence of heterozygosity (AOH) analysis demonstrated a 7.2 Mb AOH block surrounding the variant consistent with a founder allele. Two of the four affected individuals had unexplained renal failure which has not been reported in HSAN2B cases to date. Therefore, this report describes a novel RETREG1 founder allele and suggests renal failure may be an unrecognized feature of the RETREG1-disease spectrum.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Osteomyelitis , Renal Insufficiency , Alleles , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Osteomyelitis/genetics , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL