Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 211
Filter
1.
J Neuromuscul Dis ; 11(4): 735-747, 2024.
Article in English | MEDLINE | ID: mdl-38788085

ABSTRACT

Motor neuron diseases and peripheral neuropathies are heterogeneous groups of neurodegenerative disorders that manifest with distinct symptoms due to progressive dysfunction or loss of specific neuronal subpopulations during different stages of development. A few monogenic, neurodegenerative diseases associated with primary metabolic disruptions of sphingolipid biosynthesis have been recently discovered. Sphingolipids are a subclass of lipids that form critical building blocks of all cellular and subcellular organelle membranes including the membrane components of the nervous system cells. They are especially abundant within the lipid portion of myelin. In this review, we will focus on our current understanding of disease phenotypes in three monogenic, neuromuscular diseases associated with pathogenic variants in components of serine palmitoyltransferase, the first step in sphingolipid biosynthesis. These include hereditary sensory and autonomic neuropathy type 1 (HSAN1), a sensory predominant peripheral neuropathy, and two neurodegenerative disorders: juvenile amyotrophic lateral sclerosis affecting the upper and lower motor neurons with sparing of sensory neurons, and a complicated form of hereditary spastic paraplegia with selective involvement of the upper motor neurons and more broad CNS neurodegeneration. We will also review our current understanding of disease pathomechanisms, therapeutic approaches, and the unanswered questions to explore in future studies.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies , Neurodegenerative Diseases , Neurodevelopmental Disorders , Serine C-Palmitoyltransferase , Humans , Serine C-Palmitoyltransferase/metabolism , Serine C-Palmitoyltransferase/genetics , Neurodegenerative Diseases/metabolism , Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/metabolism , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Sphingolipids/metabolism
2.
Pharmacol Res ; 169: 105662, 2021 07.
Article in English | MEDLINE | ID: mdl-34000361

ABSTRACT

Nerve growth factor (NGF), by binding to TrkA and p75NTR receptors, regulates the survival and differentiation of sensory neurons during development and mediates pain transmission and perception during adulthood, by acting at different levels of the nervous system. Key to understanding the role of NGF as a pain mediator is the finding that mutations (namely, R121W, V232fs and R221W) in the NGF gene cause painlessness disease Hereditary Sensory and Autonomic Neuropathy type V (HSAN V). Here we shall review the consequences of these NGF mutations, each of which results in specific clinical signs: R221W determines congenital pain insensitivity with no overt cognitive disabilities, whereas V232fs and R121W also result in intellectual disability, thus showing similarities to HSAN IV, which is caused by mutations in TrkA, rather than to HSAN V. Comparing the cellular, biochemical and clinical findings of these mutations could help in better understanding not only the possible mechanisms underlying HSAN V, but also mechanisms of NGF signalling and roles. These mutations alter the balance between NGF and proNGF in favour of an accumulation of the latter, suggesting a possible role of proNGF as a molecule with an analgesic role. Furthermore, the neurotrophic and pronociceptive functions of NGF are split by the R221W mutation, making NGF variants based on this mutation interesting for designing therapeutic applications for many diseases. This review emphasizes the possibility of using the mutations involved in "painlessness" clinical disorders as an innovative approach to identify new proteins and pathways involved in pain transmission and perception. OUTSTANDING QUESTIONS: Why do homozygous HSAN V die postnatally? What is the cause of this early postnatal lethality? Is the development of a mouse or a human feeling less pain affecting higher cognitive and perceptual functions? What is the consequence of the HSAN V mutation on the development of joints and bones? Are the multiple fractures observed in HSAN V patients due exclusively to the carelessness consequent to not feeling pain, or also to an intrinsic frailty of their bones? Are heterodimers of NGFWT and NGFR221W in the heterozygote state formed? And if so, what are the properties of these heterodimeric proteins? How is the processing of proNGFR221W to NGFR221W affected by the mutation?


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/genetics , Nerve Growth Factor/physiology , Pain Perception/physiology , Protein Precursors/physiology , Animals , Hereditary Sensory and Autonomic Neuropathies/metabolism , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Humans , Mutation/genetics , Nerve Growth Factor/genetics , Protein Precursors/genetics
3.
Brain ; 144(8): 2457-2470, 2021 09 04.
Article in English | MEDLINE | ID: mdl-33751098

ABSTRACT

Sensory neuronopathies are a rare and distinct subgroup of peripheral neuropathies, characterized by degeneration of the dorsal root ganglia neurons. About 50% of sensory neuronopathies are idiopathic and genetic causes remain to be clarified. Through a combination of homozygosity mapping and whole exome sequencing, we linked an autosomal recessive sensory neuronopathy to pathogenic variants in the COX20 gene. We identified eight unrelated families from the eastern Chinese population carrying a founder variant c.41A>G (p.Lys14Arg) within COX20 in either a homozygous or compound heterozygous state. All patients displayed sensory ataxia with a decrease in non-length-dependent sensory potentials. COX20 encodes a key transmembrane protein implicated in the assembly of mitochondrial complex IV. We showed that COX20 variants lead to reduction of COX20 protein in patient's fibroblasts and transfected cell lines, consistent with a loss-of-function mechanism. Knockdown of COX20 expression in ND7/23 sensory neuron cells resulted in complex IV deficiency and perturbed assembly of complex IV, which subsequently compromised cell spare respiratory capacity and reduced cell proliferation under metabolic stress. Consistent with mitochondrial dysfunction in knockdown cells, reduced complex IV assembly, enzyme activity and oxygen consumption rate were also found in patients' fibroblasts. We speculated that the mechanism of COX20 was similar to other causative genes (e.g. SURF1, COX6A1, COA3 and SCO2) for peripheral neuropathies, all of which are functionally important in the structure and assembly of complex IV. Our study identifies a novel causative gene for the autosomal recessive sensory neuronopathy, whose vital function in complex IV and high expression in the proprioceptive sensory neuron further underlines loss of COX20 contributing to mitochondrial bioenergetic dysfunction as a mechanism in peripheral sensory neuron disease.


Subject(s)
Cytochrome-c Oxidase Deficiency/genetics , Electron Transport Complex IV/genetics , Hereditary Sensory and Autonomic Neuropathies/genetics , Loss of Heterozygosity , Mitochondria/genetics , Adolescent , Adult , Cell Proliferation/genetics , Child , Child, Preschool , Cytochrome-c Oxidase Deficiency/physiopathology , Female , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Humans , Male , Median Nerve/physiopathology , Mutation , Neural Conduction/physiology , Pedigree , Radial Nerve/physiopathology , Ulnar Nerve/physiopathology
4.
Prog Neurobiol ; 194: 101886, 2020 11.
Article in English | MEDLINE | ID: mdl-32693191

ABSTRACT

The R100W mutation in nerve growth factor is associated with hereditary sensory autonomic neuropathy V in a Swedish family. These patients develop severe loss of perception to deep pain but with apparently normal cognitive functions. To better understand the disease mechanism, we examined a knockin mouse model of HSAN V. The homozygous mice showed significant structural deficits in intra-epidermal nerve fibers (IENFs) at birth. These mice had a total loss of pain perception at ∼2 months of age and often failed to survive to adulthood. Heterozygous mutant mice developed a progressive degeneration of small sensory fibers both behaviorally and functionally: they showed a progressive loss of IENFs starting at the age of 9 months accompanied with progressive loss of perception to painful stimuli such as noxious temperature. Quantitative analysis of lumbar 4/5 dorsal root ganglia revealed a significant reduction in small size neurons, while analysis of sciatic nerve fibers revealed the heterozygous mutant mice had no reduction in myelinated nerve fibers. Significantly, the amount of NGF secreted from mouse embryonic fibroblasts were reduced from both heterozygous and homozygous mice compared to their wild-type littermates. Interestingly, the heterozygous mice showed no apparent structural alteration in the brain: neither the anterior cingulate cortex nor the medial septum including NGF-dependent basal forebrain cholinergic neurons. Accordingly, these animals did not develop appreciable deficits in tests for brain function. Our study has thus demonstrated that the NGFR100W mutation likely affects the structure and function of peripheral sensory neurons.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/pathology , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Nerve Growth Factor/genetics , Pain Perception/physiology , Animals , Behavior, Animal/physiology , Disease Models, Animal , Disease Progression , Embryo, Mammalian , Fibroblasts , Heterozygote , Homozygote , Learning/physiology , Mice , Mice, Transgenic , Mutation, Missense , Point Mutation , Social Behavior
5.
Ann Clin Transl Neurol ; 7(9): 1502-1512, 2020 09.
Article in English | MEDLINE | ID: mdl-32730653

ABSTRACT

OBJECTIVE: Sphingolipids are enriched in the nerves. Serine-palmitoyltransferase (SPT) catalyzes the key step of sphingolipids biosynthesis. Mutations in SPT subunits (SPTLC) lead to the excessive production of neurotoxic deoxysphingolipids (DoxSLs) in patients with Hereditary Sensory Neuropathy Type-1C (HSN1C). HSN1C is an autosomal dominant peripheral neuropathy characterized by sensory loss and distal muscle weakness. In this study, by leveraging a HSN1C family with a previously reported N177D mutation in SPTLC2, we aim to further define the spectrum of DoxSL species and the peripheral neve pathology of the disease. METHODS: Next-generation sequencing along with Sanger confirmation was performed for family members and healthy controls. LC-MS was used for lipidomic analysis in participants' plasma. Quantitative magnetic resonance imaging (qMRI) was performed to study sciatic nerve pathologies. RESULTS: A heterozygous N177D mutation in SPTLC2 was co-segregated in individuals with sensory-motor deficits in the limbs. Nerve conduction studies (NCS) revealed nonuniform slowing of conduction velocities. In line with the NCS, qMRI detected a pattern of nerve changes similar to those in acquired demyelinating polyneuropathies. Additionally, we detected a significant increase in multiple species of deoxysphingoid bases and deoxyceramides in patients' plasma. INTERPRETATION: Mutations in the SPTLC2 cause a demyelinating phenotype resembling those in acquired demyelinating polyneuropathy. The species of increased DoxSLs in HSN1C may be more diverse than originally thought.


Subject(s)
Demyelinating Diseases/diagnosis , Hereditary Sensory and Autonomic Neuropathies/diagnosis , Sphingolipids/metabolism , Adult , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Demyelinating Diseases/physiopathology , Female , Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/metabolism , Hereditary Sensory and Autonomic Neuropathies/physiopathology , High-Throughput Nucleotide Sequencing , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neural Conduction/physiology , Pedigree , Sequence Analysis, DNA , Serine C-Palmitoyltransferase/genetics , Sphingolipids/blood
7.
J Peripher Nerv Syst ; 25(3): 308-311, 2020 09.
Article in English | MEDLINE | ID: mdl-32470188

ABSTRACT

Hereditary sensory and autonomic neuropathies (HSAN) encompass a group of peripheral nervous system disorders characterized by remarkable heterogeneity from a clinical and genetic point of view. Mutations in SPTLC1 gene are responsible for HSAN type IA, which usually starts from the second to fourth decade with axonal neuropathy, sensory loss, painless distal ulcerations, and mild autonomic features, while motor involvement usually occur later as disease progresses. Beyond the classic presentation of HSAN type IA, an exceedingly rare distinct phenotype related to SPTLC1 mutations at residue serine 331 (S331) has recently been reported, characterized by earlier onset, prominent muscular atrophy, growth retardation, oculo-skeletal abnormalities, and possible respiratory complications. In this report, we describe clinical, instrumental, and genetic aspects of a 13-year-old Sri Lankan male carrying the rare de novo p.S331Y heterozygous mutation in SPTLC1 gene found by whole exome sequencing. Patient's phenotype partly overlaps with the first case previously reported, however with some additional features not described before. This work represent the second report about this rare mutation and our findings strongly reinforce the hypothesis of a clearly distinct "S331 syndrome", thus expanding the spectrum of SPTLC1-related disorders.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Serine C-Palmitoyltransferase/genetics , Adolescent , Humans , Male , Sri Lanka , Syndrome
8.
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
10.
Clin Neurol Neurosurg ; 189: 105636, 2020 02.
Article in English | MEDLINE | ID: mdl-31841741

ABSTRACT

OBJECTIVES: To present the clinical picture, the associated complications and the genetic findings of Jordanian patients diagnosed with Congenital insensitivity to pain with anhidrosis (CIPA). PATIENTS AND METHODS: This is a retrospective study including 7 patients diagnosed with CIPA presenting to Jordan University Hospital neurology clinic between 2001 and 2017. RESULTS: Among five families, seven patients were diagnose with CIPA and followed for a period ranging from one month to 6 years. The initial symptom observed in all patients was high fever in the first few days after birth, decreased sensation to pain and decreased sweating were later noted. Poor weight gain, microcephaly and global developmental delay were present in most cases. All patients had tongue ulcerations. Fingers/toes ulcerations were present in 6/7 (86.0 %), hip joint dislocation in 3/7 (43.0 %), chronic arthritis and joint swelling in 6/7 (86.0 %), corneal ulcers in 4/7 (57.1 %) and kidney amyloidosis in 1/7 (13.0 %) of all patients. Death occurred in 4/7 (57.1 %) patients. Consanguinity was present in all families. Mutation analysis revealed three variants in NTRK1 gene. The frameshift (c.1860_1861insT; p.Pro621fs) mutation was common in our series. One patient carried a novel missense mutation (c.2170 G > A; p.Gly724Ser). The third missense mutation (C2125 G > T; p.Val709Leu) was reported in a previous study in one patient. CONCLUSION: This cohort reveals a severe CIPA phenotype necessitating thorough multidisciplinary care and follow up.


Subject(s)
Arthritis/physiopathology , Corneal Ulcer/physiopathology , Developmental Disabilities/physiopathology , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Hip Dislocation, Congenital/physiopathology , Microcephaly/physiopathology , Receptor, trkA/genetics , Skin Ulcer/physiopathology , Adolescent , Body-Weight Trajectory , Child , Child, Preschool , Female , Fingers , Frameshift Mutation , Hereditary Sensory and Autonomic Neuropathies/genetics , Humans , Infant , Infant, Newborn , Jordan , Male , Mutation , Mutation, Missense , Pedigree , Retrospective Studies , Toes , Tongue Diseases/physiopathology , Ulcer/physiopathology , Young Adult
11.
J Neurosci ; 39(49): 9702-9715, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31685654

ABSTRACT

Nerve growth factor (NGF) is a key mediator of nociception, acting during the development and differentiation of dorsal root ganglion (DRG) neurons, and on adult DRG neuron sensitization to painful stimuli. NGF also has central actions in the brain, where it regulates the phenotypic maintenance of cholinergic neurons. The physiological function of NGF as a pain mediator is altered in patients with Hereditary Sensory and Autonomic Neuropathy type V (HSAN V), caused by the 661C>T transition in the Ngf gene, resulting in the R100W missense mutation in mature NGF. Homozygous HSAN V patients present with congenital pain insensitivity, but are cognitively normal. This led us to hypothesize that the R100W mutation may differentially affect the central and peripheral actions of NGF. To test this hypothesis and provide a mechanistic basis to the HSAN V phenotype, we generated transgenic mice harboring the human 661C>T mutation in the Ngf gene and studied both males and females. We demonstrate that heterozygous NGFR100W/wt mice display impaired nociception. DRG neurons of NGFR100W/wt mice are morphologically normal, with no alteration in the different DRG subpopulations, whereas skin innervation is reduced. The NGFR100W protein has reduced capability to activate pain-specific signaling, paralleling its reduced ability to induce mechanical allodynia. Surprisingly, however, NGFR100W/wt mice, unlike heterozygous mNGF+/- mice, show no learning or memory deficits, despite a reduction in secretion and brain levels of NGF. The results exclude haploinsufficiency of NGF as a mechanistic cause for heterozygous HSAN V mice and demonstrate a specific effect of the R100W mutation on nociception.SIGNIFICANCE STATEMENT The R100W mutation in nerve growth factor (NGF) causes Hereditary Sensory and Autonomic Neuropathy type V, a rare disease characterized by impaired nociception, even in apparently clinically silent heterozygotes. For the first time, we generated and characterized heterozygous knock-in mice carrying the human R100W-mutated allele (NGFR100W/wt). Mutant mice have normal nociceptor populations, which, however, display decreased activation of pain transduction pathways. NGFR100W interferes with peripheral and central NGF bioavailability, but this does not impact on CNS function, as demonstrated by normal learning and memory, in contrast with heterozygous NGF knock-out mice. Thus, a point mutation allows neurotrophic and pronociceptive functions of NGF to be split, with interesting implications for the treatment of chronic pain.


Subject(s)
Cognition , Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Mutation/genetics , Nerve Growth Factor/genetics , Nociception , Animals , Behavior, Animal , Female , Ganglia, Spinal/pathology , Gene Knock-In Techniques , Hereditary Sensory and Autonomic Neuropathies/psychology , Humans , Male , Mice , Mice, Transgenic , Mutation, Missense/genetics , Pain Measurement , Pain Perception , Psychomotor Performance , Rats , Rats, Wistar , Skin/innervation
12.
J Peripher Nerv Syst ; 24(4): 354-358, 2019 12.
Article in English | MEDLINE | ID: mdl-31596031

ABSTRACT

Hereditary sensory autonomic neuropathy (HSAN) type II is a rare, autosomal recessive, and early onset sensory neuropathy, characterized by severe and progressive sensation impairment, leading to ulcero-mutilating complications. FAM134B gene, also known as RETREG1 gene, mutations have been reported to be associated to HSAN-IIB. We report four patients from two unrelated families who developed during childhood a sensory axonal neuropathy with variable severity and pronounced nociception impairment. Complications such as recurrent ulcerations, osteomyelitis, and osteonecrosis leading to distal amputation were noticed. Dysautonomia was mild or even absent in our group of patients. Additionally, either clinical or neurophysiological motor impairment was not uncommon. Presence of upper motor neuron signs was also a distinctive feature in two related patients. After extensive workup, two novel homozygous mutations in the FAM134B gene were identified. This report expands the clinical and genetic spectrum of HSAN type II and emphasizes the phenotype variability even within the same family.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Adult , Consanguinity , Female , Humans , Mutation , Pedigree , Siblings , Young Adult
13.
Mol Biol Cell ; 30(22): 2814-2826, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31509475

ABSTRACT

Hereditary sensory and autonomic neuropathy (HSAN) types IA and IC (IA/C) are caused by elevated levels of an atypical class of lipid named 1-deoxysphingolipid (DoxSL). How elevated levels of DoxSL perturb the physiology of the cell and how the perturbations lead to HSAN IA/C are largely unknown. In this study, we show that C26-1-deoxydihydroceramide (C26-DoxDHCer) is highly toxic to the cell, while C16- and C18-DoxDHCer are less toxic. Genome-wide genetic screens and lipidomics revealed the dynamics of DoxSL accumulation and DoxSL species responsible for the toxicity over the course of DoxSL accumulation. Moreover, we show that disruption of F-actin organization, alteration of mitochondrial shape, and accumulation of hydrophobic bodies by DoxSL are not sufficient to cause complete cellular failure. We found that cell death coincides with collapsed ER membrane, although we cannot rule out other possible causes of cell death. Thus, we have unraveled key principles of DoxSL cytotoxicity that may help to explain the clinical features of HSAN IA/C.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/metabolism , Sphingolipids/metabolism , Actins/metabolism , Ceramides/toxicity , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Lipid Metabolism , Lipidomics , Lipids , Mitochondria/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Sphingolipids/genetics
14.
Sci Adv ; 5(7): eaaw1297, 2019 07.
Article in English | MEDLINE | ID: mdl-31281886

ABSTRACT

The canonical view is that touch is signaled by fast-conducting, thickly myelinated afferents, whereas pain is signaled by slow-conducting, thinly myelinated ("fast" pain) or unmyelinated ("slow" pain) afferents. While other mammals have thickly myelinated afferents signaling pain (ultrafast nociceptors), these have not been demonstrated in humans. Here, we performed single-unit axonal recordings (microneurography) from cutaneous mechanoreceptive afferents in healthy participants. We identified A-fiber high-threshold mechanoreceptors (A-HTMRs) that were insensitive to gentle touch, encoded noxious skin indentations, and displayed conduction velocities similar to A-fiber low-threshold mechanoreceptors. Intraneural electrical stimulation of single ultrafast A-HTMRs evoked painful percepts. Testing in patients with selective deafferentation revealed impaired pain judgments to graded mechanical stimuli only when thickly myelinated fibers were absent. This function was preserved in patients with a loss-of-function mutation in mechanotransduction channel PIEZO2. These findings demonstrate that human mechanical pain does not require PIEZO2 and can be signaled by fast-conducting, thickly myelinated afferents.


Subject(s)
Mechanotransduction, Cellular/physiology , Nociceptors/metabolism , Pain/metabolism , Skin/metabolism , Adult , Aged , Aged, 80 and over , Axons/physiology , Electric Stimulation , Female , Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Humans , Ion Channels/genetics , Loss of Function Mutation , Male , Mechanoreceptors/metabolism , Middle Aged , Nerve Fibers, Myelinated/metabolism , Neurons, Afferent/metabolism , Neurophysiology/methods , Signal Transduction , Young Adult
15.
Amyloid ; 26(3): 103-111, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31339362

ABSTRACT

Timely diagnosis of hereditary variant transthyretin (ATTRv) amyloidosis is critical for appropriate treatment and optimal outcomes. Significant differences are seen between patients receiving treatment and those who are not, though disease progression may continue despite treatment in some patients. Healthcare professionals caring for patients with ATTRv amyloidosis therefore need reliable ongoing assessments to understand the continuing course of disease and make appropriate treatment choices on an individual basis. Various signs and symptoms experienced by patients may be evaluated as indicators of disease progression, though there is currently no validated score that can be used for such ongoing assessment. Recognizing this situation, a group of clinicians highly experienced in ATTR amyloidosis developed an approach to understand and define disease progression in diagnosed and treated patients with ATTRv amyloidosis. The suggested approach is based on the recognition of distinct phenotypes which may usefully inform the particular tools, tests and investigations that are most likely to be appropriate for individual patients. It is aimed at implementing appropriate and ongoing assessment of patients being treated for ATTRv amyloidosis, such that the effectiveness of management can be usefully assessed throughout the course of disease and management can be tailored according to the patient's requirements.


Subject(s)
Amyloid Neuropathies, Familial/diagnosis , Cardiomyopathies/diagnosis , Disease Management , Glaucoma/diagnosis , Hereditary Sensory and Autonomic Neuropathies/diagnosis , Adult , Age of Onset , Aged , Amyloid Neuropathies, Familial/drug therapy , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/physiopathology , Cardiomyopathies/drug therapy , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Consensus , Disease Progression , Female , Glaucoma/drug therapy , Glaucoma/genetics , Glaucoma/physiopathology , Heart Function Tests , Hereditary Sensory and Autonomic Neuropathies/drug therapy , Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Humans , Kidney Function Tests , Male , Middle Aged , Mutation , Neuroprotective Agents/therapeutic use , Prealbumin/deficiency , Prealbumin/genetics
17.
J Peripher Nerv Syst ; 24(2): 224-229, 2019 06.
Article in English | MEDLINE | ID: mdl-30866134

ABSTRACT

Hereditary sensory and autonomic neuropathy type I (HSAN-1) is an autosomal dominant sensory neuropathy occurring secondary to mutations in the SPTLC1 and SPTLC2 genes. We present two generations of a single family with Ser384Phe mutation in the SPTLC2 gene located on chromosome 14q24 characterized by a typical HSAN-1c presentation, with additional findings upper motor neuron signs, early demyelinating features on nerve conduction studies, and type II juxtafoveal retinal telangiectasias also known as macular telangiectasias (MacTel II). Although HSAN1 is characterized as an axonal neuropathy, demyelinating features were identified in two subjects on serial nerve conduction studies comprising motor conduction block, temporal dispersion, and prolongation of F-waves. MacTell II is a rare syndrome characterized by bilateral macular depigmentation and Müller cell loss. It has a presumed genetic basis, and these cases suggest that the accumulation of toxic sphingoplipids may lead to Müller cell degeneration, subsequent neuronal loss, depigmentation, and progressive central macular thinning.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/genetics , Motor Neurons/physiology , Mutation , Retinal Telangiectasis/genetics , Serine C-Palmitoyltransferase/genetics , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Humans , Male , Middle Aged , Neural Conduction/genetics , Pedigree , Retinal Telangiectasis/physiopathology
18.
J Peripher Nerv Syst ; 24(1): 150-155, 2019 03.
Article in English | MEDLINE | ID: mdl-30680846

ABSTRACT

Hereditary sensory neuropathy (HSN) comprises a group of progressive peripheral neuropathies predominantly affecting the sensory nerves. To date, two different ATL3 gene mutations have been reported to be responsible for HSN type 1F (HSN1F). Here, we report a family in which the members presented numbness of the lower limbs and recurrent foot ulceration. Symptoms of foot ulcers disappeared in the years after onset, which suggests that the family members showed benign and mild symptoms compared with the affected patients reported previously. Laboratory examinations and electrophysiological data suggested axonal degeneration of the peripheral sensory nerves, while motor neurons were not involved. Exome sequencing revealed the previously reported c.C1013G (p.Pro338Arg) mutation of the ATL3 gene. This is the first report of ATL3 mutation in Chinese patients with HSN. Cells expressing mutant ATL3 exhibited disruption of the endoplasmic reticulum network, suggesting a dominant-negative effect. There was no significant difference in the expression of the endoplasmic reticulum stress marker binding immunoglobulin protein (BiP) between cells expressing wild-type or mutant ATL3. Further studies are required to ascertain the relevance of the changes in endoplasmic reticulum morphology to axonal degeneration of sensory nerves.


Subject(s)
Endoplasmic Reticulum/genetics , GTP Phosphohydrolases/genetics , Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Adult , China , Female , Humans , Male , Mutation , Pedigree
19.
Neurosci Lett ; 702: 51-60, 2019 05 29.
Article in English | MEDLINE | ID: mdl-30503913

ABSTRACT

Over the last 20 years a large number of transgenic mouse models have been produced showing different degrees of congenital hypoalgesia; some of these models mimic known human conditions while others seemingly have no human counterpart. However, very little significant contributions to our understanding of pain neurobiology were obtained from this multitude of animal models; in most cases the study of these animals was limited to the characterization of its pain perception without addressing the long term consequences of their hypoalgesic condition. In this review we discuss the untapped potential that these animal models of congenital hypoalgesia hold for future studies addressing brain plasticity during permanent conditions of reduced pain perception, and that may result in important insights on the interplay between pain, emotion, and cognition. Revisiting hypoalgesia using modern techniques of functional neurophysiology in awake animals may complement the recent literature of functional clinical and preclinical studies that improve our understanding of the central malplasticity caused by pain.


Subject(s)
Disease Models, Animal , Pain Perception , Pain/congenital , Animals , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Hereditary Sensory and Autonomic Neuropathies/psychology , Humans , Mice , Mice, Transgenic , Pain/physiopathology , Pain/psychology
20.
Rev. chil. neuropsicol. (En línea) ; 13(2): 46-51, dic. 2018. tab, ilus
Article in Spanish | LILACS | ID: biblio-1100633

ABSTRACT

La neuropatía sensitiva autonómica hereditaria tipo IV (HSAN-IV) es una condición neurológica de origen genético extremadamente rara que puede cursar con discapacidad intelectual, sin embargo, hay escasas publicaciones sobre las características del funcionamiento cognitivo global y la conducta adaptativa de los afectados. En este estudio se describe la capacidad cognitiva global y el funcionamiento adaptativo de dos niñas de 12 y 14 años diagnosticadas con HSANIV, incluyendo una caracterización de los procesos de comprensión verbal, razonamiento perceptual, memoria de trabajo y velocidad de procesamiento. Las menores fueron evaluadas mediante la Escala de Inteligencia para niños de Wechsler cuarta edición (WISC-IV) encontrándose en ambos casos un bajo índice de comprensión verbal, una medida del desarrollo cognitivo alcanzado a través de la historia de aprendizaje de las niñas; así como un bajo índice de razonamiento perceptivo, indicador de su capacidad para adaptarse y afrontar situaciones nuevas de forma flexible. Esto se acompaña de dificultad en la manipulación de información en la memoria para la resolución de problemas y enlentecimiento en la velocidad de procesamiento de la información. Adicionalmente, se evaluó su funcionamiento adaptativo mediante el sistema de evaluación de la conducta adaptativa ABAS-II, el cual se caracterizó por fortalezas en habilidades comunicativas, uso de recursos comunitarios y vida en el hogar; con limitaciones en habilidades académicas y de autocuidado. En conclusión, la HSAN-IV es una condición que cursa con discapacidad intelectual con necesidades de apoyo variables en intensidad. En los casos estudiados se encontró discapacidad intelectual con necesidad de apoyo limitado, es decir, los apoyos se requieren de forma regular durante un periodo de tiempo corto pero definido.


Hereditary sensory autonomic neuropathy type IV (HSAN-IV) is a neurological condition of extremely rare genetic origin that may be associated with intellectual disability; however, there are few publications about the characteristics of global cognitive functioning and adaptive behaviour of these patients. In this study we describe the global cognitive function and the adaptive behavior of two girls aged 12 and 14 diagnosed with HSAN-IV, including a characterization of the processes of verbal comprehension, perceptual reasoning, working memory and processing speed. The children were assessed using the Wechsler Intelligence Scale for Children - Fourth Edition (WISC-IV), finding in both cases a low level of verbal comprehension, a measure of cognitive development achieved through the girls' learning history; as well as a low rate of perceptual reasoning, indicator of their ability to adapt and face new situations in a flexible way. This is accompanied by difficulty in manipulating information in the memory to solve problems and slow down the speed of information processing. Additionally, its adaptive functioning was evaluated through the Adaptive Behavior Assessment System ABAS-II, which was characterized by strengths in communication skills, use of community resources and life at home; with limitations in academic and self-care skills. In conclusion, HSAN-IV is a condition related with intellectual disability with varying support needs in intensity. In the cases studied, intellectual disability was found with limited need for support, that is, supports are required on a regular basis for a short but defined period.


Subject(s)
Humans , Female , Child , Adolescent , Adaptation, Psychological/physiology , Hereditary Sensory and Motor Neuropathy/psychology , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Cognition/physiology , Refsum Disease
SELECTION OF CITATIONS
SEARCH DETAIL