Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
J Lipid Res ; 62: 100122, 2021.
Article in English | MEDLINE | ID: mdl-34563520

ABSTRACT

Hereditary sensory neuropathy type 1 (HSAN1) is a rare axonopathy, characterized by a progressive loss of sensation (pain, temperature, and vibration), neuropathic pain, and wound healing defects. HSAN1 is caused by several missense mutations in the serine palmitoyltransferase long-chain base subunit 1 and serine palmitoyltransferase long-chain base subunit 2 of the enzyme serine palmitoyltransferase-the key enzyme for the synthesis of sphingolipids. The mutations change the substrate specificity of serine palmitoyltransferase, which then forms an atypical class of 1-deoxy-sphinglipids (1-deoxySLs). Similarly, patients with type 2 diabetes mellitus also present with elevated 1-deoxySLs and a comparable clinical phenotype. The effect of 1-deoxySLs on neuronal cells was investigated in detail, but their impact on other cell types remains elusive. Here, we investigated the consequences of externally added 1-deoxySLs on the migration of fibroblasts in a scratch assay as a simplified cellular wound-healing model. We showed that 1-deoxy-sphinganine (1-deoxySA) inhibits the migration of NIH-3T3 fibroblasts in a dose- and time-dependent manner. This was not seen for a non-native, L-threo stereoisomer. Supplemented 1-deoxySA was metabolized to 1-deoxy-(dihydro)ceramide and downstream to 1-deoxy-sphingosine. Inhibiting downstream metabolism by blocking N-acylation rescued the migration phenotype. In contrast, adding 1-deoxy-sphingosine had a lesser effect on cell migration but caused the massive formation of intracellular vacuoles. Further experiments showed that the effect on cell migration was primarily mediated by 1-deoxy-dihydroceramides rather than by the free base or 1-deoxyceramides. Based on these findings, we suggest that limiting the N-acylation of 1-deoxySA could be a therapeutic approach to improve cell migration and wound healing in patients with HSAN1 and type 2 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Fibroblasts/drug effects , Hereditary Sensory and Autonomic Neuropathies/metabolism , Sphingolipids/pharmacology , Animals , Cell Movement/drug effects , Cells, Cultured , Fibroblasts/metabolism , Mice , NIH 3T3 Cells
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.
Biochim Biophys Acta Biomembr ; 1863(8): 183628, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33915167

ABSTRACT

1-deoxy-sphingolipids, also known as atypical sphingolipids, are directly implicated in the development and progression of hereditary sensory and autonomic neuropathy type 1 and diabetes type 2. The mechanisms underlying their patho-physiological actions are yet to be elucidated. Accumulating evidence suggests that the biological actions of canonical sphingolipids are triggered by changes promoted on membrane organization and biophysical properties. However, little is known regarding the biophysical implications of atypical sphingolipids. In this study, we performed a comprehensive characterization of the effects of the naturally occurring 1-deoxy-dihydroceramide, 1-deoxy-ceramideΔ14Z and 1-deoxymethyl-ceramideΔ3E in the properties of a fluid membrane. In addition, to better define which structural features determine sphingolipid ability to form ordered domains, the synthetic 1-O-methyl-ceramideΔ4E and 1-deoxy-ceramideΔ4E were also studied. Our results show that natural and synthetic 1-deoxy(methyl)-sphingolipids fail to laterally segregate into ordered domains as efficiently as the canonical C16-ceramide. The impaired ability of atypical sphingolipids to form ordered domains was more dependent on the presence, position, and configuration of the sphingoid base double bond than on the structure of its C1 functional group, due to packing constraints introduced by an unsaturated backbone. Nonetheless, absence of a hydrogen bond donor and acceptor group at the C1 position strongly reduced the capacity of atypical sphingolipids to form gel domains. Altogether, the results showed that 1-deoxy(methyl)-sphingolipids induce unique changes on the biophysical properties of the membranes, suggesting that these alterations might, in part, trigger the patho-biological actions of these lipids.


Subject(s)
Ceramides/chemistry , Lipids/chemistry , Membranes/chemistry , Sphingolipids/chemistry , Biophysics , Ceramides/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/metabolism , Hereditary Sensory and Autonomic Neuropathies/pathology , Humans , Membranes/metabolism , Sphingolipids/metabolism
4.
Clin Nucl Med ; 46(3): 227-228, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33351513

ABSTRACT

ABSTRACT: A 30-year-old man presented with repeated episodes of painless injuries in his feet and abnormally high body temperature. He was diagnosed with congenital insensitivity to pain with anhidrosis-a rare hereditary peripheral neuropathy characterized by decreased pain, reduced sweating, and autonomic neuropathy. Congenital insensitivity to pain with anhidrosis is also called hereditary sensory and autonomic neuropathy type IV. 123I-MIBG myocardial scintigraphy showed reduced myocardial uptake (heart-to-mediastinum ratio: 1.56 and 1.42 in the early and late phases, respectively; washout ratio, 49%), indicating autonomic dysfunction. This finding may contribute to the diagnosis of congenital insensitivity to pain with anhidrosis and the semiquantitative evaluation of an autonomic dysfunction.


Subject(s)
3-Iodobenzylguanidine/metabolism , Hereditary Sensory and Autonomic Neuropathies/metabolism , Myocardium/metabolism , Adult , Biological Transport , Hereditary Sensory and Autonomic Neuropathies/diagnostic imaging , Humans , Male , Myocardial Perfusion Imaging
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
6.
Cell Physiol Biochem ; 54(4): 577-590, 2020 Jun 08.
Article in English | MEDLINE | ID: mdl-32506846

ABSTRACT

BACKGROUND/AIMS: Loss-of-Function (LOF) of the potassium chloride cotransporter 3 (KCC3) results in hereditary sensorimotor neuropathy with Agenesis of the Corpus Callosum (HSMN/ACC). Our KCC3 knockout mouse recapitulated axonal swelling and tissue vacuolization observed in autopsies of individuals with HSMN/ACC. We previously documented the first human case of a KCC3 gain-of-function (GOF) in which the patient also exhibited severe peripheral neuropathy. Furthermore, the GOF mouse model exhibited shrunken axons implicating the cotransporter in cell volume homeostasis. It is unclear how both KCC3 LOF and GOF lead to peripheral neuropathy. Thus, we sought to study differences in cell volume regulation of dorsal root ganglion neurons isolated from different mouse lines. METHODS: Using wide-field microscopy, we measured calcein fluorescence intensity through pinhole measurements at the center of cells and compared cell swelling and cell volume regulation/recovery of wild-type, LOF, and GOF dorsal root ganglia neurons, as well as wild-type neurons treated with a KCC-specific inhibitor. RESULTS: In contrast to control neurons that swell and volume regulate under a hypotonic challenge, neurons lacking KCC3 swell but fail to volume regulate. Similar data were observed in wild-type neurons treated with the KCC inhibitor. We also show that sensory neurons expressing a constitutively active KCC3 exhibited a blunted swelling phase compared to wild-type neurons, questioning the purely osmotic nature of the swelling phase. CONCLUSION: These findings demonstrate the integral role of KCC3 in cell volume homeostasis and support the idea that cell volume homeostasis is critical to the health of peripheral nerves.


Subject(s)
Corpus Callosum/metabolism , Ganglia, Spinal/metabolism , Hereditary Sensory and Autonomic Neuropathies/metabolism , Membrane Transport Proteins/metabolism , Neurons/metabolism , Symporters/metabolism , Animals , Axons/metabolism , Cell Size/drug effects , Corpus Callosum/pathology , Disease Models, Animal , Gain of Function Mutation , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Hereditary Sensory and Autonomic Neuropathies/genetics , Homeostasis , Humans , Mice , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Osmotic Pressure/physiology , Symporters/antagonists & inhibitors , Symporters/genetics
7.
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
8.
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
9.
J Neurosci ; 39(29): 5816-5834, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31138658

ABSTRACT

Hereditary sensory and autonomic neuropathy Type 1 (HSAN1) is a rare autosomal dominantly inherited neuropathy, clinically characterized by a loss of distal peripheral sensory and motoneuronal function. Mutations in subunits of serine palmitoyltransferase (SPT) have been linked to the majority of HSAN1 cases. SPTs catalyze the condensation of l-serine with palmitoyl-CoA, the first committed and rate-limiting step in de novo sphingolipid biosynthesis. Despite extensive investigation, the molecular pathogenesis of HSAN1 remains controversial. Here, we established a Caenorhabditis elegans (C. elegans) model of HSAN1 by generating a sptl-1(c363g) mutation, encoding SPTL-1(C121W) and equivalent to human SPTLC1C133W, at the C. elegans genomic locus through CRISPR. The sptl-1(c363g) homozygous mutants exhibited the same larval lethality and epithelial polarity defect as observed in sptl-1(RNAi) animals, suggesting a loss-of-function effect of the SPTL-1(C121W) mutation. sptl-1(c363g)/+ heterozygous mutants displayed sensory dysfunction with concomitant neuronal morphology and axon-dendrite polarity defects, demonstrating that the C. elegans model recapitulates characteristics of the human disease. sptl-1(c363g)-derived neuronal defects were copied in animals with defective sphingolipid biosynthetic enzymes downstream of SPTL-1, including ceramide glucosyltransferases, suggesting that SPTLC1C133W contributes to the HSAN1 pathogenesis by limiting the production of complex sphingolipids, including glucosylceramide. Overexpression of SPTL-1(C121W) led to similar epithelial and neuronal defects and to reduced levels of complex sphingolipids, specifically glucosylceramide, consistent with a dominant-negative effect of SPTL-1(C121W) that is mediated by loss of this downstream product. Genetic interactions between SPTL-1(C121W) and components of directional trafficking in neurons suggest that the neuronal polarity phenotype could be caused by glycosphingolipid-dependent defects in polarized vesicular trafficking.SIGNIFICANCE STATEMENT The symptoms of inherited metabolic diseases are often attributed to the accumulation of toxic intermediates or byproducts, no matter whether the disease-causing enzyme participates in a biosynthetic or a degradation pathway. By showing that the phenotypes observed in a C. elegans model of HSAN1 disease could be caused by loss of a downstream product (glucosylceramide) rather than the accumulation of a toxic byproduct, our work provides new insights into the origins of the symptoms of inherited metabolic diseases while expanding the repertoire of sphingolipid functions, specifically, of glucosylceramides. These findings not only have their most immediate relevance for neuroprotective treatments for HSAN1, they may also have implications for a much broader range of neurologic conditions.


Subject(s)
Cell Polarity/physiology , Disease Models, Animal , Glycosphingolipids/metabolism , Hereditary Sensory and Autonomic Neuropathies/metabolism , Neurons/physiology , Animals , Animals, Genetically Modified , Base Sequence , Caenorhabditis elegans , Glycosphingolipids/genetics , Hereditary Sensory and Autonomic Neuropathies/genetics , Humans
10.
BMC Med Genet ; 20(1): 91, 2019 05 27.
Article in English | MEDLINE | ID: mdl-31132985

ABSTRACT

BACKGROUND: Hereditary sensory and autonomic neuropathy (HSAN) type II is a group of extremely rare autosomal recessive neurological disorders with heterogeneous clinical and genetic characteristics. METHODS: We performed high-depth next-generation targeted sequencing using a custom-ordered "HSAN" panel, covering WNK1, NTRK1, NGF, SPTLC1 and IKBKAP genes, to identify pathogenic variants of the proband as well as the family members. We also performed whole exome sequencing to further investigate the potential occurrence of additional pathogenic variants in genes that were not covered by the "HSAN" panel. Quantitative real-time PCR was used to identify pathogenic copy number variations (CNVs) and to analyze the mRNA level of WNK1 gene of the family. Western blot analysis was performed to evaluate the WNK1 protein expression level. RESULTS: After sequencing, a novel nonsense variant (c.2747 T > G, p.Leu916Ter) in exon 9 of WNK1 gene was identified in two patients (hemizygous) and their mother (heterozygous). This variant is absent in all public databases as well as in 600 Han Chinese healthy controls. The region of this variant is evolutionary highly conserved. Furthermore, by quantitative real-time PCR using DNA of the pedigree, we revealed a large deletion containing the whole WNK1 gene in two patients. The WNK1 expression levels of the patients were significantly reduced. CONCLUSIONS: Our study firstly revealed that the coexistence of a novel WNK1 nonsense variant and a CNV resulted in HSAN type IIA in a Han Chinese family.


Subject(s)
Codon, Nonsense , DNA Copy Number Variations , Hereditary Sensory and Autonomic Neuropathies/genetics , WNK Lysine-Deficient Protein Kinase 1/genetics , Amino Acid Sequence , Base Sequence , DNA Mutational Analysis , Family Health , Female , Hereditary Sensory and Autonomic Neuropathies/metabolism , Hereditary Sensory and Autonomic Neuropathies/pathology , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Pedigree , Sequence Homology, Amino Acid , WNK Lysine-Deficient Protein Kinase 1/metabolism
11.
Hum Mol Genet ; 28(4): 615-627, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30339187

ABSTRACT

Axonopathies are neurodegenerative disorders caused by axonal degeneration, affecting predominantly the longest neurons. Several of these axonopathies are caused by genetic defects in proteins involved in the shaping and dynamics of the endoplasmic reticulum (ER); however, it is unclear how these defects impinge on neuronal survival. Given its central and widespread position within a cell, the ER is a pivotal player in inter-organelle communication. Here, we demonstrate that defects in the ER fusion protein ATL3, which were identified in patients suffering from hereditary sensory and autonomic neuropathy, result in an increased number of ER-mitochondria contact sites both in HeLa cells and in patient-derived fibroblasts. This increased contact is reflected in higher phospholipid metabolism, upregulated autophagy and augmented Ca2+ crosstalk between both organelles. Moreover, the mitochondria in these cells display lowered motility, and the number of axonal mitochondria in neurons expressing disease-causing mutations in ATL3 is strongly decreased. These results underscore the functional interdependence of subcellular organelles in health and disease and show that disorders caused by ER-shaping defects are more complex than previously assumed.


Subject(s)
Axons/metabolism , Endoplasmic Reticulum/genetics , GTP Phosphohydrolases/genetics , Hereditary Sensory and Autonomic Neuropathies/genetics , Autophagy/genetics , Axons/pathology , Calcium/metabolism , Calcium Signaling/genetics , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , HeLa Cells , Hereditary Sensory and Autonomic Neuropathies/metabolism , Hereditary Sensory and Autonomic Neuropathies/pathology , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mutation , Neurons/metabolism , Neurons/pathology
12.
Mol Pain ; 14: 1744806918809223, 2018.
Article in English | MEDLINE | ID: mdl-30296891

ABSTRACT

Bi-allelic dysfunctional mutations in nerve growth factor (NGF) cause the rare human phenotype hereditary sensory and autonomic neuropathy type 5 (HSAN5). We describe a novel NGF mutation in an individual with typical HSAN5 findings. The mutation c.361C>T, p.R121W is at the last residue of the furin cleavage motif Arg-Ser-Lys-Arg in proNGF. We show that the p.R121W mutation completely abolishes the formation of mature NGF-ß. Surprisingly, mutant p.R121W cells produced very little proNGF. Instead, the two progressive cleavage products of proNGF were produced, proA-NGF and proB-NGF, with proB-NGF being the predominant NGF-derived peptide and the only peptide secreted by mutant p.R121W cells. We found that the ability of the p.R121W mutation to cause tropomyosin receptor kinase A autophosphorylation and mitogen-activated protein kinase phosphorylation was significantly reduced compared to controls (p < 0.05 and p < 0.01). By studying the PC12 cell line morphology and neurite length over a week, we found the p.R121W mutation had residual, but much reduced, neurotrophic activity when compared to wild-type NGF. Finally, we assessed whether the p.R121W mutation affected apoptosis and found a reduced protective effect compared to wild-type NGF. Our results suggest that the p.R121W NGF mutation causes HSAN5 through negating the ability of furin to cleave proNGF to produce NGF-ß.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/genetics , Mutation/genetics , Nerve Growth Factor/metabolism , Neurites/metabolism , Neurons/metabolism , Animals , Hereditary Sensory and Autonomic Neuropathies/metabolism , PC12 Cells , Phosphorylation/genetics , Protein Precursors/metabolism , Rats
13.
Biochem J ; 475(19): 3009-3034, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30291170

ABSTRACT

Actin is folded to its native state in eukaryotic cytosol by the sequential allosteric mechanism of the chaperonin-containing TCP-1 (CCT). The CCT machine is a double-ring ATPase built from eight related subunits, CCT1-CCT8. Non-native actin interacts with specific subunits and is annealed slowly through sequential binding and hydrolysis of ATP around and across the ring system. CCT releases a folded but soft ATP-G-actin monomer which is trapped 80 kJ/mol uphill on the folding energy surface by its ATP-Mg2+/Ca2+ clasp. The energy landscape can be re-explored in the actin filament, F-actin, because ATP hydrolysis produces dehydrated and more compact ADP-actin monomers which, upon application of force and strain, are opened and closed like the elements of a spring. Actin-based myosin motor systems underpin a multitude of force generation processes in cells and muscles. We propose that the water surface of F-actin acts as a low-binding energy, directional waveguide which is recognized specifically by the myosin lever-arm domain before the system engages to form the tight-binding actomyosin complex. Such a water-mediated recognition process between actin and myosin would enable symmetry breaking through fast, low energy initial binding events. The origin of chaperonins and the subsequent emergence of the CCT-actin system in LECA (last eukaryotic common ancestor) point to the critical role of CCT in facilitating phagocytosis during early eukaryotic evolution and the transition from the bacterial world. The coupling of CCT-folding fluxes to the cell cycle, cell size control networks and cancer are discussed together with directions for further research.


Subject(s)
Actins/metabolism , Chaperonin Containing TCP-1/metabolism , Evolution, Molecular , Protein Folding , Actins/chemistry , Actins/genetics , Amino Acid Sequence , Animals , Chaperonin Containing TCP-1/chemistry , Chaperonin Containing TCP-1/genetics , Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/metabolism , Hereditary Sensory and Autonomic Neuropathies/pathology , Humans , Mutation/genetics , Protein Structure, Secondary , Protein Structure, Tertiary
14.
Hum Mol Genet ; 27(20): 3598-3611, 2018 10 15.
Article in English | MEDLINE | ID: mdl-29982604

ABSTRACT

Hereditary sensory and autonomic neuropathy type VI (HSAN-VI) is a recessive human disease that arises from mutations in the dystonin gene (DST; also known as Bullous pemphigoid antigen 1 gene). A milder form of HSAN-VI was recently described, resulting from loss of a single dystonin isoform (DST-A2). Similarly, mutations in the mouse dystonin gene (Dst) result in severe sensory neuropathy, dystonia musculorum (Dstdt). Two Dstdt alleles, Dstdt-Tg4 and Dstdt-27J, differ in the severity of disease. The less severe Dstdt-Tg4 mice have disrupted expression of Dst-A1 and -A2 isoforms, while the more severe Dstdt-27J allele affects Dst-A1, -A2 and -A3 isoforms. As dystonin is a cytoskeletal-linker protein, we evaluated microtubule network integrity within sensory neurons from Dstdt-Tg4 and Dstdt-27J mice. There is a significant reduction in tubulin acetylation in Dstdt-27J indicative of microtubule instability and severe microtubule disorganization within sensory axons. However, Dstdt-Tg4 mice have no change in tubulin acetylation, and microtubule organization was only mildly impaired. Thus, microtubule instability is not central to initiation of Dstdt pathogenesis, though it may contribute to disease severity. Maintenance of microtubule stability in Dstdt-Tg4 dorsal root ganglia could be attributed to an upregulation in Dst-A3 expression as a compensation for the absence of Dst-A1 and -A2 in Dstdt-Tg4 sensory neurons. Indeed, knockdown of Dst-A3 in these neurons resulted in a decrease in tubulin acetylation. These findings shed light on the possible compensatory role of dystonin isoforms within HSAN-VI, which might explain the heterogeneity in symptoms within the reported forms of the disease.


Subject(s)
Dystonic Disorders/genetics , Dystonin/genetics , Gene Expression Regulation , Hereditary Sensory and Autonomic Neuropathies/genetics , Protein Processing, Post-Translational , Tubulin/metabolism , Acetylation , Animals , Cell Line , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Dystonic Disorders/metabolism , Dystonin/metabolism , Hereditary Sensory and Autonomic Neuropathies/metabolism , Mice , Microtubules/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Protein Isoforms , Up-Regulation
15.
Neurobiol Dis ; 117: 1-14, 2018 09.
Article in English | MEDLINE | ID: mdl-29778900

ABSTRACT

Hereditary sensory neuropathy type 1 (HSN-1) is a peripheral neuropathy most frequently caused by mutations in the SPTLC1 or SPTLC2 genes, which code for two subunits of the enzyme serine palmitoyltransferase (SPT). SPT catalyzes the first step of de novo sphingolipid synthesis. Mutations in SPT result in a change in enzyme substrate specificity, which causes the production of atypical deoxysphinganine and deoxymethylsphinganine, rather than the normal enzyme product, sphinganine. Levels of these abnormal compounds are elevated in blood of HSN-1 patients and this is thought to cause the peripheral motor and sensory nerve damage that is characteristic of the disease, by a largely unresolved mechanism. In this study, we show that exogenous application of these deoxysphingoid bases causes dose- and time-dependent neurotoxicity in primary mammalian neurons, as determined by analysis of cell survival and neurite length. Acutely, deoxysphingoid base neurotoxicity manifests in abnormal Ca2+ handling by the endoplasmic reticulum (ER) and mitochondria as well as dysregulation of cell membrane store-operated Ca2+ channels. The changes in intracellular Ca2+ handling are accompanied by an early loss of mitochondrial membrane potential in deoxysphingoid base-treated motor and sensory neurons. Thus, these results suggest that exogenous deoxysphingoid base application causes neuronal mitochondrial dysfunction and Ca2+ handling deficits, which may play a critical role in the pathogenesis of HSN-1.


Subject(s)
Calcium/metabolism , Hereditary Sensory and Autonomic Neuropathies/metabolism , Mitochondria/metabolism , Motor Neurons/metabolism , Sphingolipids/toxicity , Animals , Cells, Cultured , Hereditary Sensory and Autonomic Neuropathies/pathology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/pathology , Motor Neurons/drug effects , Motor Neurons/pathology
16.
J Neurosci ; 38(14): 3394-3413, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29483280

ABSTRACT

Nerve growth factor (NGF) exerts multiple functions on target neurons throughout development. The recent discovery of a point mutation leading to a change from arginine to tryptophan at residue 100 in the mature NGFß sequence (NGFR100W) in patients with hereditary sensory and autonomic neuropathy type V (HSAN V) made it possible to distinguish the signaling mechanisms that lead to two functionally different outcomes of NGF: trophic versus nociceptive. We performed extensive biochemical, cellular, and live-imaging experiments to examine the binding and signaling properties of NGFR100W Our results show that, similar to the wild-type NGF (wtNGF), the naturally occurring NGFR100W mutant was capable of binding to and activating the TrkA receptor and its downstream signaling pathways to support neuronal survival and differentiation. However, NGFR100W failed to bind and stimulate the 75 kDa neurotrophic factor receptor (p75NTR)-mediated signaling cascades (i.e., the RhoA-Cofilin pathway). Intraplantar injection of NGFR100W into adult rats induced neither TrkA-mediated thermal nor mechanical acute hyperalgesia, but retained the ability to induce chronic hyperalgesia based on agonism for TrkA signaling. Together, our studies provide evidence that NGFR100W retains trophic support capability through TrkA and one aspect of its nociceptive signaling, but fails to engage p75NTR signaling pathways. Our findings suggest that wtNGF acts via TrkA to regulate the delayed priming of nociceptive responses. The integration of both TrkA and p75NTR signaling thus appears to regulate neuroplastic effects of NGF in peripheral nociception.SIGNIFICANCE STATEMENT In the present study, we characterized the naturally occurring nerve growth factor NGFR100W mutant that is associated with hereditary sensory and autonomic neuropathy type V. We have demonstrated for the first time that NGFR100W retains trophic support capability through TrkA, but fails to engage p75NTR signaling pathways. Furthermore, after intraplantar injection into adult rats, NGFR100W induced neither thermal nor mechanical acute hyperalgesia, but retained the ability to induce chronic hyperalgesia. We have also provided evidence that the integration of both TrkA- and p75NTR-mediated signaling appears to regulate neuroplastic effects of NGF in peripheral nociception. Our study with NGFR100W suggests that it is possible to uncouple trophic effect from nociceptive function, both induced by wild-type NGF.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/genetics , Mutation, Missense , Nerve Growth Factor/genetics , Nociception , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/metabolism , 3T3 Cells , Animals , Cells, Cultured , HEK293 Cells , Hereditary Sensory and Autonomic Neuropathies/metabolism , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Humans , Male , Mice , Nerve Growth Factor/metabolism , Nerve Tissue Proteins , PC12 Cells , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, Growth Factor , Signal Transduction
17.
J Cell Physiol ; 233(6): 4479-4489, 2018 06.
Article in English | MEDLINE | ID: mdl-29226326

ABSTRACT

FAM134B (family with sequence similarity 134, member B)/RETREG1 and its functional roles are relatively new in human diseases. This review aimed to summarize various functions of FAM134B since our first discovery of the gene in 2001. The protein encoded by FAM134B is a reticulophagy receptor that regulates turnover of the endoplasmic reticulum (ER) by selective phagocytosis. Absence or non-functional expression of FAM134B protein impairs ER-turnover and thereby is involved in the pathogenesis of some human diseases. FAM134B inhibition contributes to impair proteostasis in the ER due to the accumulation of misfolded or aggregated proteins, which in turn leads to compromised neuronal survival and progressive neuronal degenerative diseases. Mutations in FAM134B associated with hereditary sensory and autonomic neuropathy type IIB (HSAN IIB). Selective cleavage of FAM134B by Dengue, Zika, and West Nile virus encoded protease NS2B3 leads to the increased production of infection units, whereas upregulation of FAM134B inhibits viral replication. In cancer, FAM134B acts as a tumor suppressor and inhibit cancer growth both in-vitro and in-vivo. Pharmacological upregulation of FAM134B resulted in reduced cancer cell growth and proliferation. In addition, FAM134B mutations are common in patients with colorectal adenocarcinoma, and oesophageal squamous cell carcinoma. These mutations and expression changes of FAM134B were associated with the biological aggressiveness of these cancers. FAM134B also plays a role in allergic rhinitis, vascular dementia, and identification of stem cells. Taken together, information available in the literature suggests that FAM134B plays critical roles in human diseases, by interacting with different biological and chemical mediators, which are primarily regulated by ER turnover.


Subject(s)
Biomarkers, Tumor/metabolism , Hereditary Sensory and Autonomic Neuropathies/metabolism , Inflammation/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Vascular Diseases/metabolism , Virus Diseases/metabolism , Animals , Autophagy , Biomarkers, Tumor/genetics , Endoplasmic Reticulum Stress , Gene Expression Regulation, Neoplastic , Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/physiopathology , Humans , Inflammation/genetics , Inflammation/physiopathology , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/physiopathology , Signal Transduction , Vascular Diseases/genetics , Vascular Diseases/physiopathology , Virus Diseases/genetics , Virus Diseases/virology
18.
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
19.
Hum Mutat ; 38(1): 55-63, 2017 01.
Article in English | MEDLINE | ID: mdl-27676246

ABSTRACT

Hereditary sensory and autonomic neuropathy type IV (HSAN IV) is an autosomal recessive disorder characterized by a complete lack of pain perception and anhidrosis. Here, we studied a cohort of seven patients with HSAN IV and describe a comprehensive functional analysis of seven novel NTRK1 missense mutations, c.1550G >A, c.1565G >A, c.1970T >C, c.2096T >C, c.2254T >A, c.2288G >C, and c.2311C >T, corresponding to p.G517E, p.G522E, p.L657P, p.I699T, p.C752S, p.C763S, and p.R771C, all of which were predicted pathogenic by in silico analysis. The results allowed us to assess the pathogenicity of each mutation and to gain novel insights into tropomyosin receptor kinase A (TRKA) downstream signaling. Each mutation was systematically analyzed for TRKA glycosylation states, intracellular and cell membrane expression patterns, nerve growth factor stimulated TRKA autophosphorylation, TRKA-Y496 phosphorylation, PLCγ activity, and neurite outgrowth. We showed a diverse range of functional effects: one mutation appeared fully functional, another had partial activity in all assays, one mutation affected only the PLCγ pathway and four mutations were proved null in all assays. Thus, we conclude that complete abolition of TRKA kinase activity is not the only pathogenic mechanism underlying HSAN IV. By corollary, the assessment of the clinical pathogenicity of HSAN IV mutations is more complex than initially predicted and requires a multifaceted approach.


Subject(s)
Hereditary Sensory and Autonomic Neuropathies/genetics , Hereditary Sensory and Autonomic Neuropathies/metabolism , Mutation, Missense , Receptor, trkA/genetics , Receptor, trkA/metabolism , Alleles , Cell Line , Computational Biology/methods , DNA Mutational Analysis , Gene Order , Genetic Association Studies , Genetic Loci , Genetic Predisposition to Disease , Genotype , Glycosylation , Hereditary Sensory and Autonomic Neuropathies/diagnosis , Humans , Molecular Imaging , Neurites/metabolism , Phospholipase C gamma/metabolism , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Receptor, trkA/chemistry , Recombinant Fusion Proteins , Sequence Analysis, DNA , Signal Transduction
20.
J Lipid Res ; 58(1): 60-71, 2017 01.
Article in English | MEDLINE | ID: mdl-27872144

ABSTRACT

The 1-deoxysphingolipids (1-deoxySLs) are atypical sphingolipids (SLs) that are formed when serine palmitoyltransferase condenses palmitoyl-CoA with alanine instead of serine during SL synthesis. The 1-deoxySLs are toxic to neurons and pancreatic ß-cells. Pathologically elevated 1-deoxySLs cause the inherited neuropathy, hereditary sensory autonomic neuropathy type 1 (HSAN1), and are also found in T2D. Diabetic sensory polyneuropathy (DSN) and HSAN1 are clinically very similar, suggesting that 1-deoxySLs may be implicated in both pathologies. The 1-deoxySLs are considered to be dead-end metabolites, as they lack the C1-hydroxyl group, which is essential for the canonical degradation of SLs. Here, we report a previously unknown metabolic pathway, which is capable of degrading 1-deoxySLs. Using a variety of metabolic labeling approaches and high-resolution high-accuracy MS, we identified eight 1-deoxySL downstream metabolites, which appear to be formed by cytochrome P450 (CYP)4F enzymes. Comprehensive inhibition and induction of CYP4F enzymes blocked and stimulated, respectively, the formation of the downstream metabolites. Consequently, CYP4F enzymes might be novel therapeutic targets for the treatment of HSAN1 and DSN, as well as for the prevention of T2D.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Diabetic Neuropathies/metabolism , Hereditary Sensory and Autonomic Neuropathies/metabolism , Sphingolipids/metabolism , Animals , Cytochrome P-450 Enzyme System/genetics , Diabetic Neuropathies/genetics , Diabetic Neuropathies/pathology , Hereditary Sensory and Autonomic Neuropathies/pathology , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Metabolic Networks and Pathways/genetics , Mice , Mutation , Oxidation-Reduction , Serine C-Palmitoyltransferase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...