Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 397
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 87: 351-390, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29195049

ABSTRACT

In this review, we describe speculative ideas and early stage research concerning the flow of genetic information from the nuclear residence of genes to the disparate, cytoplasmic sites of protein synthesis. We propose that this process of information transfer is meticulously guided by transient structures formed from protein segments of low sequence complexity/intrinsic disorder. These low complexity domains are ubiquitously associated with regulatory proteins that control gene expression and RNA biogenesis, but they are also found in the central channel of nuclear pores, the nexus points of intermediate filament assembly, and the locations of action of other well-studied cellular proteins and pathways. Upon being organized into localized cellular positions via mechanisms utilizing properly folded protein domains, thereby facilitating elevated local concentration, certain low complexity domains adopt cross-ß interactions that are both structurally specific and labile to disassembly. These weakly tethered assemblies, we propose, are built to relay the passage of genetic information from one site to another within a cell, ensuring that the process is of extreme fidelity.


Subject(s)
Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Models, Biological , Animals , Cytoplasmic Granules/genetics , Cytoplasmic Granules/metabolism , Gene Expression , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/metabolism , Humans , Hydrogels , Intrinsically Disordered Proteins/chemistry , Models, Molecular , Mutation , Protein Interaction Domains and Motifs , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Protein FUS/chemistry , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism
2.
Mol Cell ; 75(5): 1073-1085.e6, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31327635

ABSTRACT

Mitochondrial AAA+ quality-control proteases regulate diverse aspects of mitochondrial biology through specialized protein degradation, but the underlying mechanisms of these enzymes remain poorly defined. The mitochondrial AAA+ protease AFG3L2 is of particular interest, as genetic mutations localized throughout AFG3L2 are linked to diverse neurodegenerative disorders. However, a lack of structural data has limited our understanding of how mutations impact enzymatic function. Here, we used cryoelectron microscopy (cryo-EM) to determine a substrate-bound structure of the catalytic core of human AFG3L2. This structure identifies multiple specialized structural features that integrate with conserved motifs required for ATP-dependent translocation to unfold and degrade targeted proteins. Many disease-relevant mutations localize to these unique structural features of AFG3L2 and distinctly influence its activity and stability. Our results provide a molecular basis for neurological phenotypes associated with different AFG3L2 mutations and establish a structural framework to understand how different members of the AAA+ superfamily achieve specialized biological functions.


Subject(s)
ATP-Dependent Proteases/chemistry , ATPases Associated with Diverse Cellular Activities/chemistry , Mitochondrial Proteins/chemistry , Mutation , ATP-Dependent Proteases/genetics , ATP-Dependent Proteases/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Cryoelectron Microscopy , HEK293 Cells , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/metabolism , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Protein Domains
4.
Hum Mol Genet ; 29(22): 3616-3630, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33215680

ABSTRACT

Spastic paraplegia 35 (SPG35) (OMIM: 612319) or fatty acid hydroxylase-associated neurodegeneration (FAHN) is caused by deficiency of fatty acid 2-hydroxylase (FA2H). This enzyme synthesizes sphingolipids containing 2-hydroxylated fatty acids, which are particularly abundant in myelin. Fa2h-deficient (Fa2h-/-) mice develop symptoms reminiscent of the human disease and therefore serve as animal model of SPG35. In order to understand further the pathogenesis of SPG35, we compared the proteome of purified CNS myelin isolated from wild type and Fa2h-/- mice at different time points of disease progression using tandem mass tag labeling. Data analysis with a focus on myelin membrane proteins revealed a significant increase of the oligodendrocytic myelin paranodal and inner loop protein (Opalin) in Fa2h-/- mice, whereas the concentration of other major myelin proteins was not significantly changed. Western blot analysis revealed an almost 6-fold increase of Opalin in myelin of Fa2h-/- mice aged 21-23 months. A concurrent unaltered Opalin gene expression suggested a decreased turnover of the Opalin protein in Fa2h-/- mice. Supporting this hypothesis, Opalin protein half-life was reduced significantly when expressed in CHO cells synthesizing 2-hydroxylated sulfatide, compared to cells synthesizing only non-hydroxylated sulfatide. Degradation of Opalin was inhibited by inhibitors of lysosomal degradation but unaffected by proteasome inhibitors. Taken together, these results reveal a new function of 2-hydroxylated sphingolipids namely affecting the turnover of a myelin membrane protein. This may play a role in the pathogenesis of SPG35.


Subject(s)
Amidohydrolases/genetics , Heredodegenerative Disorders, Nervous System/genetics , Myelin Proteins/genetics , Myelin Sheath/genetics , Spastic Paraplegia, Hereditary/genetics , Animals , Disease Models, Animal , Gene Expression Regulation/genetics , Heredodegenerative Disorders, Nervous System/metabolism , Heredodegenerative Disorders, Nervous System/pathology , Humans , Mice , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Pedigree , Spastic Paraplegia, Hereditary/metabolism , Spastic Paraplegia, Hereditary/pathology , Sphingolipids/biosynthesis , Sphingolipids/genetics
5.
Neurol Sci ; 44(12): 4359-4362, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37410270

ABSTRACT

BACKGROUND: NBIA (neurodegeneration with brain iron accumulation) is a diverse collection of neurodegenerative illnesses defined by iron accumulation in the basal ganglia. The fatty acid hydroxylase-associated neurodegeneration, or FAHN, is one of the uncommon subtypes of NBIAs, associated with inherited autosomal recessive mutations in gene coding the membrane-bound fatty acid 2 hydroxylase (FA2H) enzyme. CASES: Here, we report two cases with FAHN from two unrelated families from Iran confirmed by whole exome sequencing. CONCLUSION: FAHN is an uncommon variant of NBIA that may manifest as spastic paraparesis without signs of iron buildup on brain imaging. As a result, it should be taken into account while making a differential diagnosis of the hereditary spastic paraplegia (HSP) syndrome, especially in individuals who lack iron deposits.


Subject(s)
Heredodegenerative Disorders, Nervous System , Pantothenate Kinase-Associated Neurodegeneration , Spastic Paraplegia, Hereditary , Humans , Brain/diagnostic imaging , Heredodegenerative Disorders, Nervous System/genetics , Iran , Iron , Mutation/genetics , Pantothenate Kinase-Associated Neurodegeneration/genetics , Spastic Paraplegia, Hereditary/diagnostic imaging , Spastic Paraplegia, Hereditary/genetics
6.
J Neurochem ; 158(2): 217-232, 2021 07.
Article in English | MEDLINE | ID: mdl-33864399

ABSTRACT

Gangliosides are expressed on plasma membranes throughout the body and enriched in the nervous system. A critical role for complex a- and b-series gangliosides in central and peripheral nervous system ageing has been established through transgenic manipulation of enzymes in ganglioside biosynthesis. Disrupting GalNAc-transferase (GalNAc-T), thus eliminating all a- and b-series complex gangliosides (with consequent over-expression of GM3 and GD3) leads to an age-dependent neurodegeneration. Mice that express only GM3 ganglioside (double knockout produced by crossing GalNAc-T-/- and GD3 synthase-/- mice, Dbl KO) display markedly accelerated neurodegeneration with reduced survival. Degenerating axons and disrupted node of Ranvier architecture are key features of complex ganglioside-deficient mice. Previously, we have shown that reintroduction of both a- and b-series gangliosides into neurons on a global GalNAcT-/- background is sufficient to rescue this age-dependent neurodegenerative phenotype. To determine the relative roles of a- and b-series gangliosides in this rescue paradigm, we herein reintroduced GalNAc-T into neurons of Dbl KO mice, thereby reconstituting a-series but not b-series complex gangliosides. We assessed survival, axon degeneration, axo-glial integrity, inflammatory markers and lipid-raft formation in these Rescue mice compared to wild-type and Dbl KO mice. We found that this neuronal reconstitution of a-series complex gangliosides abrogated the adult lethal phenotype in Dbl KO mice, and partially attenuated the neurodegenerative features. This suggests that whilst neuronal expression of a-series gangliosides is critical for survival during ageing, it is not entirely sufficient to restore complete nervous system integrity in the absence of either b-series or glial a-series gangliosides.


Subject(s)
G(M3) Ganglioside/metabolism , Gangliosides/metabolism , Genes, Lethal/genetics , Neurons/metabolism , Animals , Axons/pathology , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/pathology , Inflammation/metabolism , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , N-Acetylgalactosaminyltransferases/genetics , Phenotype , Ranvier's Nodes/pathology , Sialyltransferases/genetics , Survival Analysis , Polypeptide N-acetylgalactosaminyltransferase
7.
Brain ; 143(6): 1811-1825, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32436573

ABSTRACT

The polyglutamine (polyQ) diseases are a group of inherited neurodegenerative diseases that include Huntington's disease, various spinocerebellar ataxias, spinal and bulbar muscular atrophy, and dentatorubral pallidoluysian atrophy. They are caused by the abnormal expansion of a CAG repeat coding for the polyQ stretch in the causative gene of each disease. The expanded polyQ stretches trigger abnormal ß-sheet conformational transition and oligomerization followed by aggregation of the polyQ proteins in the affected neurons, leading to neuronal toxicity and neurodegeneration. Disease-modifying therapies that attenuate both symptoms and molecular pathogenesis of polyQ diseases remain an unmet clinical need. Here we identified arginine, a chemical chaperone that facilitates proper protein folding, as a novel compound that targets the upstream processes of polyQ protein aggregation by stabilizing the polyQ protein conformation. We first screened representative chemical chaperones using an in vitro polyQ aggregation assay, and identified arginine as a potent polyQ aggregation inhibitor. Our in vitro and cellular assays revealed that arginine exerts its anti-aggregation property by inhibiting the toxic ß-sheet conformational transition and oligomerization of polyQ proteins before the formation of insoluble aggregates. Arginine exhibited therapeutic effects on neurological symptoms and protein aggregation pathology in Caenorhabditis elegans, Drosophila, and two different mouse models of polyQ diseases. Arginine was also effective in a polyQ mouse model when administered after symptom onset. As arginine has been safely used for urea cycle defects and for mitochondrial myopathy, encephalopathy, lactic acid and stroke syndrome patients, and efficiently crosses the blood-brain barrier, a drug-repositioning approach for arginine would enable prompt clinical application as a promising disease-modifier drug for the polyQ diseases.


Subject(s)
Arginine/metabolism , Arginine/pharmacology , Peptides/metabolism , Animals , Caenorhabditis elegans/metabolism , Disease Models, Animal , Drosophila/metabolism , Female , Heredodegenerative Disorders, Nervous System/genetics , Huntington Disease/genetics , Male , Mice , Mice, Inbred Strains , Molecular Chaperones/genetics , Peptides/genetics , Protein Aggregation, Pathological , Protein Conformation/drug effects , Protein Folding/drug effects , Spinocerebellar Ataxias/genetics
8.
Brain ; 143(1): 94-111, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31855247

ABSTRACT

Cerebral choline metabolism is crucial for normal brain function, and its homoeostasis depends on carrier-mediated transport. Here, we report on four individuals from three families with neurodegenerative disease and homozygous frameshift mutations (Asp517Metfs*19, Ser126Metfs*8, and Lys90Metfs*18) in the SLC44A1 gene encoding choline transporter-like protein 1. Clinical features included progressive ataxia, tremor, cognitive decline, dysphagia, optic atrophy, dysarthria, as well as urinary and bowel incontinence. Brain MRI demonstrated cerebellar atrophy and leukoencephalopathy. Moreover, low signal intensity in globus pallidus with hyperintensive streaking and low signal intensity in substantia nigra were seen in two individuals. The Asp517Metfs*19 and Ser126Metfs*8 fibroblasts were structurally and functionally indistinguishable. The most prominent ultrastructural changes of the mutant fibroblasts were reduced presence of free ribosomes, the appearance of elongated endoplasmic reticulum and strikingly increased number of mitochondria and small vesicles. When chronically treated with choline, those characteristics disappeared and mutant ultrastructure resembled healthy control cells. Functional analysis revealed diminished choline transport yet the membrane phosphatidylcholine content remained unchanged. As part of the mechanism to preserve choline and phosphatidylcholine, choline transporter deficiency was implicated in impaired membrane homeostasis of other phospholipids. Choline treatments could restore the membrane lipids, repair cellular organelles and protect mutant cells from acute iron overload. In conclusion, we describe a novel childhood-onset neurometabolic disease caused by choline transporter deficiency with autosomal recessive inheritance.


Subject(s)
Antigens, CD/genetics , Heredodegenerative Disorders, Nervous System/genetics , Organic Cation Transport Proteins/genetics , Adolescent , Ataxia/genetics , Ataxia/physiopathology , Atrophy , Cerebellum/diagnostic imaging , Cerebellum/pathology , Choline/pharmacology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/physiopathology , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/ultrastructure , Deglutition Disorders/genetics , Deglutition Disorders/physiopathology , Dysarthria/genetics , Dysarthria/physiopathology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/ultrastructure , Fecal Incontinence/genetics , Fecal Incontinence/physiopathology , Female , Fibroblasts/drug effects , Fibroblasts/ultrastructure , Frameshift Mutation , Globus Pallidus/diagnostic imaging , Heredodegenerative Disorders, Nervous System/diagnostic imaging , Heredodegenerative Disorders, Nervous System/pathology , Heredodegenerative Disorders, Nervous System/physiopathology , Homozygote , Humans , Leukoencephalopathies/diagnostic imaging , Leukoencephalopathies/genetics , Leukoencephalopathies/physiopathology , Magnetic Resonance Imaging , Male , Microscopy, Electron , Mitochondria/drug effects , Mitochondria/ultrastructure , Nootropic Agents/pharmacology , Optic Atrophy/genetics , Optic Atrophy/physiopathology , Pedigree , Ribosomes/drug effects , Ribosomes/ultrastructure , Substantia Nigra/diagnostic imaging , Syndrome , Tremor/genetics , Tremor/physiopathology , Urinary Incontinence/genetics , Urinary Incontinence/physiopathology
9.
Int J Mol Sci ; 22(22)2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34830106

ABSTRACT

Mitochondrial DNA depletion syndromes (MDS) are clinically heterogenous and often severe diseases, characterized by a reduction of the number of copies of mitochondrial DNA (mtDNA) in affected tissues. In the context of MDS, yeast has proved to be both an excellent model for the study of the mechanisms underlying mitochondrial pathologies and for the discovery of new therapies via high-throughput assays. Among the several genes involved in MDS, it has been shown that recessive mutations in MPV17 cause a hepatocerebral form of MDS and Navajo neurohepatopathy. MPV17 encodes a non selective channel in the inner mitochondrial membrane, but its physiological role and the nature of its cargo remains elusive. In this study we identify ten drugs active against MPV17 disorder, modelled in yeast using the homologous gene SYM1. All ten of the identified molecules cause a concomitant increase of both the mitochondrial deoxyribonucleoside triphosphate (mtdNTP) pool and mtDNA stability, which suggests that the reduced availability of DNA synthesis precursors is the cause for the mtDNA deletion and depletion associated with Sym1 deficiency. We finally evaluated the effect of these molecules on mtDNA stability in two other MDS yeast models, extending the potential use of these drugs to a wider range of MDS patients.


Subject(s)
DNA, Fungal , DNA, Mitochondrial , Heredodegenerative Disorders, Nervous System , Liver Diseases , Membrane Proteins , Mitochondria , Mitochondrial Diseases , Mitochondrial Proteins , Peripheral Nervous System Diseases , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , DNA, Fungal/genetics , DNA, Fungal/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/metabolism , Heredodegenerative Disorders, Nervous System/therapy , Humans , Liver Diseases/genetics , Liver Diseases/metabolism , Liver Diseases/therapy , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/therapy , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nucleotides/genetics , Nucleotides/metabolism , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/therapy , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Syndrome
10.
Pediatr Allergy Immunol ; 31 Suppl 24: 5-7, 2020 02.
Article in English | MEDLINE | ID: mdl-32017225

ABSTRACT

Gene therapy has become promising in many fields of medicine, as a single treatment could allow long-lasting and curative benefits. New medicines based on cell gene correction are expected to occur in upcoming years and will be hopefully part of the therapeutic armamentarium for inherited and acquired diseases. Issues related to the costs of these new therapies and access to care for all patients, and procedures and expertise needed to facilitate their application worldwide require to be addressed, together with long-term safety and efficacy monitoring.


Subject(s)
Genetic Diseases, Inborn/therapy , Genetic Therapy/methods , Heredodegenerative Disorders, Nervous System/therapy , Immune System Diseases/therapy , Zinc Finger Nucleases/metabolism , Animals , CRISPR-Cas Systems , Gene Editing , Genetic Diseases, Inborn/genetics , Genetic Therapy/trends , Genetic Vectors , Heredodegenerative Disorders, Nervous System/genetics , Humans , Immune System Diseases/genetics , Zinc Finger Nucleases/genetics
11.
Brain ; 142(8): 2230-2237, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31332433

ABSTRACT

Superoxide dismutase 1 (SOD1) is the principal cytoplasmic superoxide dismutase in humans and plays a major role in redox potential regulation. It catalyses the transformation of the superoxide anion (O2•-) into hydrogen peroxide. Heterozygous variants in SOD1 are a common cause of familial amyotrophic lateral sclerosis. In this study we describe the homozygous truncating variant c.335dupG (p.C112Wfs*11) in SOD1 that leads to total absence of enzyme activity. The resulting phenotype is severe and marked by progressive loss of motor abilities, tetraspasticity with predominance in the lower extremities, mild cerebellar atrophy, and hyperekplexia-like symptoms. Heterozygous carriers have a markedly reduced enzyme activity when compared to wild-type controls but show no overt neurologic phenotype. These results are in contrast with the previously proposed theory that a loss of function is the underlying mechanism in SOD1-related motor neuron disease and should be considered before application of previously proposed SOD1 silencing as a treatment option for amyotrophic lateral sclerosis.


Subject(s)
Heredodegenerative Disorders, Nervous System/genetics , Superoxide Dismutase-1/deficiency , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis , Child , Child, Preschool , Frameshift Mutation , Humans , Male , Pedigree , Syndrome
12.
Brain ; 142(6): 1561-1572, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31135052

ABSTRACT

The endoplasmic reticulum enzyme fatty acid 2-hydroxylase (FA2H) plays a major role in the formation of 2-hydroxy glycosphingolipids, main components of myelin. FA2H deficiency in mice leads to severe central demyelination and axon loss. In humans it has been associated with phenotypes from the neurodegeneration with brain iron accumulation (fatty acid hydroxylase-associated neurodegeneration, FAHN), hereditary spastic paraplegia (HSP type SPG35) and leukodystrophy (leukodystrophy with spasticity and dystonia) spectrum. We performed an in-depth clinical and retrospective neurophysiological and imaging study in a cohort of 19 cases with biallelic FA2H mutations. FAHN/SPG35 manifests with early childhood onset predominantly lower limb spastic tetraparesis and truncal instability, dysarthria, dysphagia, cerebellar ataxia, and cognitive deficits, often accompanied by exotropia and movement disorders. The disease is rapidly progressive with loss of ambulation after a median of 7 years after disease onset and demonstrates little interindividual variability. The hair of FAHN/SPG35 patients shows a bristle-like appearance; scanning electron microscopy of patient hair shafts reveals deformities (longitudinal grooves) as well as plaque-like adhesions to the hair, likely caused by an abnormal sebum composition also described in a mouse model of FA2H deficiency. Characteristic imaging features of FAHN/SPG35 can be summarized by the 'WHAT' acronym: white matter changes, hypointensity of the globus pallidus, ponto-cerebellar atrophy, and thin corpus callosum. At least three of four imaging features are present in 85% of FA2H mutation carriers. Here, we report the first systematic, large cohort study in FAHN/SPG35 and determine the phenotypic spectrum, define the disease course and identify clinical and imaging biomarkers.


Subject(s)
Heredodegenerative Disorders, Nervous System/genetics , Phenotype , Spastic Paraplegia, Hereditary/genetics , Child , Cohort Studies , Demyelinating Diseases/genetics , Female , Humans , Male , Mixed Function Oxygenases/genetics , Mutation/genetics , Pedigree , Retrospective Studies , Spastic Paraplegia, Hereditary/classification
13.
Curr Genet ; 65(1): 17-28, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29974202

ABSTRACT

Trinucleotide repeats are a particular class of microsatellites whose large expansions are responsible for at least two dozen human neurological and developmental disorders. Slippage of the two complementary DNA strands during replication, homologous recombination or DNA repair is generally accepted as a mechanism leading to repeat length changes, creating expansions and contractions of the repeat tract. The present review focuses on recent developments on double-strand break repair involving trinucleotide repeat tracts. Experimental evidences in model organisms show that gene conversion and break-induced replication may lead to large repeat tract expansions, while frequent contractions occur either by single-strand annealing between repeat ends or by gene conversion, triggering near-complete contraction of the repeat tract. In the second part of this review, different therapeutic approaches using highly specific single- or double-strand endonucleases targeted to trinucleotide repeat loci are compared. Relative efficacies and specificities of these nucleases will be discussed, as well as their potential strengths and weaknesses for possible future gene therapy of these dramatic disorders.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Genetic Therapy/methods , Trinucleotide Repeats/genetics , DNA/genetics , DNA/metabolism , Endonucleases/metabolism , Genetic Therapy/trends , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/metabolism , Humans , Models, Genetic , Trinucleotide Repeat Expansion/genetics
14.
Clin Genet ; 95(1): 182-186, 2019 01.
Article in English | MEDLINE | ID: mdl-30298599

ABSTRACT

MPV17 encodes a putative channel-forming protein of the inner mitochondrial membrane and is involved in mitochondrial deoxynucleotide homeostasis. MPV17 mutations were first reported in patients with Navajo neurohepatopathy, an autosomal recessive mitochondrial DNA depletion syndrome, characterized by early-onset liver failure, failure to thrive as well as central and peripheral neurological involvement. Recently, two patients with juvenile-onset peripheral sensorimotor neuropathy associated with an MVP17 c.122G>A (p.Arg41Gln) variant have been reported. Here, we describe five additional patients from two unrelated families with sensorimotor axonal neuropathy without hepatocerebral affection caused by homozygous MPV17 variants. Patients of the first family carried the known c.122G>A variant and affected individuals of the second family had a novel c.376-9T>G near-splice variant, which was shown to result in an in-frame deletion of 11 amino acids. This report provides further evidence that MPV17 mutations should be considered in patients with pure, non-syndromic axonal neuropathy.


Subject(s)
Genetic Predisposition to Disease , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Peripheral Nervous System Diseases/genetics , Polyneuropathies/genetics , Adolescent , Adult , Age of Onset , Axons/pathology , Child , Failure to Thrive/genetics , Failure to Thrive/physiopathology , Female , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/physiopathology , Humans , Liver Diseases/genetics , Liver Diseases/physiopathology , Liver Failure/genetics , Liver Failure/physiopathology , Male , Mitochondrial Diseases/genetics , Mitochondrial Diseases/physiopathology , Peripheral Nervous System Diseases/physiopathology , Polyneuropathies/physiopathology , Sensorimotor Cortex/physiopathology , Young Adult
15.
Ann Neurol ; 83(1): 153-165, 2018 01.
Article in English | MEDLINE | ID: mdl-29283439

ABSTRACT

OBJECTIVE: Autosomal-recessive mutations in TBCK cause intellectual disability of variable severity. Although the physiological function of TBCK remains unclear, loss-of-function mutations are associated with inhibition of mechanistic target of rapamycin complex 1 (mTORC1) signaling. Given that mTORC1 signaling is known to regulate autophagy, we hypothesized that TBCK-encephalopathy patients with a neurodegenerative course have defects in autophagic-lysosomal dysfunction. METHODS: Children (n = 8) of Puerto Rican (Boricua) descent affected with homozygous TBCK p.R126X mutations underwent extensive neurological phenotyping and neurophysiological studies. We quantified autophagosome content in TBCK-/- patient-derived fibroblasts by immunostaining and assayed autophagic markers by western assay. Free sialylated oligosaccharide profiles were assayed in patient's urine and fibroblasts. RESULTS: The neurological phenotype of children with TBCK p.R126X mutations, which we call TBCK-encephaloneuronopathy (TBCKE), include congenital hypotonia, progressive motor neuronopathy, leukoencephalopathy, and epilepsy. Systemic features include coarse facies, dyslipidemia, and osteoporosis. TBCK-/- fibroblasts in vitro exhibit increased numbers of LC3+ autophagosomes and increased autophagic flux by immunoblots. Free oligosaccharide profiles in fibroblasts and urine of TBCKE patients differ from control fibroblasts and are ameliorated by treatment with the mTORC1 activator leucine. INTERPRETATION: TBCKE is a clinically distinguishable syndrome with progressive central and peripheral nervous system dysfunction, consistently observed in patients with the p.R126X mutation. We provide evidence that inappropriate autophagy in the absence of cellular stressors may play a role in this disorder, and that mTORC1 activation may ameliorate the autophagic-lysosomal system dysfunction. Free oligosaccharide profiles could serve as a novel biomarker for this disorder as well as a tool to evaluate potential therapeutic interventions. Ann Neurol 2018;83:153-165.


Subject(s)
Autophagy/genetics , Heredodegenerative Disorders, Nervous System/genetics , Mutation/genetics , Protein Serine-Threonine Kinases/genetics , Adolescent , Biomarkers/analysis , Child , Exome/genetics , Fibroblasts , Heredodegenerative Disorders, Nervous System/pathology , Humans , Intellectual Disability , Leucine/therapeutic use , Male , Mechanistic Target of Rapamycin Complex 1/agonists , Mechanistic Target of Rapamycin Complex 1/biosynthesis , Oligosaccharides/analysis , Phagosomes/pathology , Phenotype , Puerto Rico
16.
Scand J Gastroenterol ; 54(12): 1441-1447, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31814461

ABSTRACT

Background: Intestinal degenerative neuropathy without extra-intestinal involvement occurs as familial forms (FIDN) but the genetics behind is unknown. We studied a Swedish family with autosomal dominant disease and several cases of chronic intestinal pseudo-obstruction (CIP).Methods: We included 33 members of a family sharing a male ancestor. Chronic intestinal symptoms including diarrhoea occurred in 11, four had severe CIP. DNA was analysed with SNP-microarray (Affymetrix), linkage (Allegro Software) and gene dosage (CNAG 3.0).Results: Genetic linkage was found to the short arm of Ch9 to a 9.7 Mb region with 45 protein-coding genes, 22 of which were duplicated (1.2 Mb duplication) (dup(9)(p21.3) with breaking point in the FOCAD-gene. Lod score for the region was 3.4. Fourteen subjects were duplication carriers including all 11 subjects having severe chronic symptoms/CIP. Nineteen subjects had no duplication. The occurrence of gastrointestinal symptoms in the family was strongly linked to duplication carrier-ship (p = .0005). The two branches of the family had separate maternal ancestors (A and B). Including the previous generation, severe disease (overt CIP and/or death from intestinal failure) was assessed to occur in 100% (5/5) of duplication carriers in branch A and in 21% (3/14) in branch B (p = .005). In branch B the onset of symptoms was later (median 38 vs. 24 yrs) and three duplication carriers were symptom-free.Conclusions: In this family with autosomal dominant hereditary intestinal neuropathy, the disorder is linked to a 9.7 Mb region in Ch9 including a 1.2 Mb duplication. There is a significant difference in disease expressivity between family branches, seemingly related to separate maternal ancestors.


Subject(s)
Chromosomes, Human, Pair 9 , Diarrhea , Heredodegenerative Disorders, Nervous System , Intestinal Pseudo-Obstruction , Intestines , Nerve Tissue Proteins/genetics , Adult , Chronic Disease , Diarrhea/diagnosis , Diarrhea/etiology , Family , Female , Gene Duplication , Genetic Loci , Heredodegenerative Disorders, Nervous System/diagnosis , Heredodegenerative Disorders, Nervous System/epidemiology , Heredodegenerative Disorders, Nervous System/genetics , Humans , Intestinal Pseudo-Obstruction/epidemiology , Intestinal Pseudo-Obstruction/etiology , Intestinal Pseudo-Obstruction/physiopathology , Intestines/innervation , Intestines/physiopathology , Male , Pedigree , Severity of Illness Index , Sweden/epidemiology
17.
Rev Neurol (Paris) ; 175(4): 238-246, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30293881

ABSTRACT

Facial-onset sensory and motor neuronopathy (FOSMN) syndrome represents a rare, slowly progressive, lower motor neuron disease with sensory compromise, involving mainly the face, bulbar region and upper limbs. However, non-motor symptoms and neurogenetic studies have rarely been evaluated in large case series. In the present study, 10 unrelated Brazilian patients with FOSMN syndrome underwent extensive clinical, laboratory, neurophysiological and neurogenetic assessment. Median age at symptom onset was 52.1 years, and men and women were equally affected. Patients presented with hemifacial or bilateral facial paresthesia and weakness, which evolved with dysphagia, dysphonia, and facial and tongue atrophy and, finally, a dropped-head, upper limb weakness and syringomyelia-like sensory disturbances in the upper limbs. All 10 patients showed chronic diffuse neurogenic compromise of bulbar, cervical and thoracic myotomes, and abnormal blink reflex tests. A positive family history of neurodegeneration was identified in six cases, and revealed pathogenic gene variants in three families (involving VCP, TARDBP and CHCHD10). Thus, our case series has revealed new findings regarding FOSMN syndrome: (i) its clinical course is not always benign, with poorer prognoses associated with dropped-head syndrome and early bulbar compromise; (ii) FOSMN syndrome may be part of a complex familial neurodegenerative spectrum; and (iii) a definite genetic basis may be observed in some cases.


Subject(s)
Facial Nerve Diseases/physiopathology , Motor Neuron Disease/physiopathology , Adult , Age of Onset , Aged , Blinking , Brazil , Facial Nerve Diseases/diagnostic imaging , Facial Nerve Diseases/genetics , Female , Genetic Testing , Heredodegenerative Disorders, Nervous System/epidemiology , Heredodegenerative Disorders, Nervous System/genetics , Humans , Male , Middle Aged , Motor Neuron Disease/diagnostic imaging , Motor Neuron Disease/genetics , Muscle Weakness/etiology , Muscular Atrophy, Spinal/epidemiology , Neuroimaging , Neurologic Examination , Paresthesia/etiology
18.
Hum Mutat ; 39(4): 461-470, 2018 04.
Article in English | MEDLINE | ID: mdl-29282788

ABSTRACT

Mitochondrial DNA (mtDNA) maintenance defects are a group of diseases caused by deficiency of proteins involved in mtDNA synthesis, mitochondrial nucleotide supply, or mitochondrial dynamics. One of the mtDNA maintenance proteins is MPV17, which is a mitochondrial inner membrane protein involved in importing deoxynucleotides into the mitochondria. In 2006, pathogenic variants in MPV17 were first reported to cause infantile-onset hepatocerebral mtDNA depletion syndrome and Navajo neurohepatopathy. To date, 75 individuals with MPV17-related mtDNA maintenance defect have been reported with 39 different MPV17 pathogenic variants. In this report, we present an additional 25 affected individuals with nine novel MPV17 pathogenic variants. We summarize the clinical features of all 100 affected individuals and review the total 48 MPV17 pathogenic variants. The vast majority of affected individuals presented with an early-onset encephalohepatopathic disease characterized by hepatic and neurological manifestations, failure to thrive, lactic acidemia, and mtDNA depletion detected mainly in liver tissue. Rarely, MPV17 deficiency can cause a late-onset neuromyopathic disease characterized by myopathy and peripheral neuropathy with no or minimal liver involvement. Approximately half of the MPV17 pathogenic variants are missense. A genotype with biallelic missense variants, in particular homozygous p.R50Q, p.P98L, and p.R41Q, can carry a relatively better prognosis.


Subject(s)
DNA, Mitochondrial/genetics , Heredodegenerative Disorders, Nervous System , Liver Diseases , Membrane Proteins/genetics , Mitochondrial Diseases , Mitochondrial Proteins/genetics , Peripheral Nervous System Diseases , Heredodegenerative Disorders, Nervous System/diagnosis , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/metabolism , Humans , Liver/metabolism , Liver Diseases/diagnosis , Liver Diseases/genetics , Liver Diseases/metabolism , Mitochondria/genetics , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mutation , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/metabolism
19.
Hum Mol Genet ; 25(21): 4635-4648, 2016 11 01.
Article in English | MEDLINE | ID: mdl-28158450

ABSTRACT

Mutation in a growing spectrum of genes is known to either cause or contribute to primary or secondary microcephaly. In primary microcephaly the genetic determinants frequently involve mutations that contribute to or modulate the microtubule cytoskeleton by causing perturbations of neuronal proliferation and migration. Here we describe four patients from two unrelated families each with an infantile neurodegenerative disorder characterized by loss of developmental milestones at 9­24 months of age followed by seizures, dystonia and acquired microcephaly. The patients harboured homozygous missense mutations (A475T and A586V) in TBCD, a gene encoding one of five tubulin-specific chaperones (termed TBCA-E) that function in concert as a nanomachine required for the de novo assembly of the α/ß tubulin heterodimer. The latter is the subunit from which microtubule polymers are assembled. We found a reduced intracellular abundance of TBCD in patient fibroblasts to about 10% (in the case of A475T) or 40% (in the case of A586V) compared to age-matched wild type controls. Functional analyses of the mutant proteins revealed a partially compromised ability to participate in the heterodimer assembly pathway. We show via in utero shRNA-mediated suppression that a balanced supply of tbcd is critical for cortical cell proliferation and radial migration in the developing mouse brain. We conclude that TBCD is a novel functional contributor to the mammalian cerebral cortex development, and that the pathological mechanism resulting from the mutations we describe is likely to involve compromised interactions with one or more TBCD-interacting effectors that influence the dynamics and behaviour of the neuronal cytoskeleton.


Subject(s)
Heredodegenerative Disorders, Nervous System/genetics , Microcephaly/genetics , Microtubule-Associated Proteins/genetics , Animals , Brain/metabolism , Cytoskeleton/metabolism , Fibroblasts/metabolism , Heredodegenerative Disorders, Nervous System/metabolism , Humans , Infant , Infant, Newborn , Mice , Mice, Inbred C57BL/embryology , Microtubule-Associated Proteins/metabolism , Microtubules/genetics , Microtubules/physiology , Tubulin/genetics , Tubulin/metabolism , Exome Sequencing/methods
20.
Mol Pharm ; 15(12): 5781-5792, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30392378

ABSTRACT

Polyglutamine diseases are a set of progressive neurodegenerative disorders caused by misfolding and aggregation of mutant CAG RNA and polyglutamin protein. To date, there is a lack of effective therapeutics that can counteract the polyglutamine neurotoxicity. Two peptidylic inhibitors, QBP1 and P3, targeting the protein and RNA toxicities, respectively, have been previously demonstrated by us with combinational therapeutic effects on the Drosophila polyglutamine disease model. However, their therapeutic efficacy has never been investigated in vivo in mammals. The current study aims to (a) develop a brain-targeting delivery system for both QBP1 and L1P3V8 (a lipidated variant of P3 with improved stability) and (b) evaluate their therapeutic effects on the R6/2 transgenic mouse model of polyglutamine disease. Compared with intravenous administration, intranasal administration of QBP1 significantly increased its brain-to-plasma ratio. In addition, employment of a chitosan-containing in situ gel for the intranasal administration of QBP1 notably improved its brain concentration for up to 10-fold. Further study on intranasal cotreatment with the optimized formulation of QBP1 and L1P3V8 in mice found no interference on the brain uptake of each other. Subsequent efficacy evaluation of 4-week daily QBP1 (16 µmol/kg) and L1P3V8 (6 µmol/kg) intranasal cotreatment in the R6/2 mice demonstrated a significant improvement on the motor coordination and explorative behavior of the disease mice, together with a full suppression on the RNA- and protein-toxicity markers in their brains. In summary, the current study developed an efficient intranasal cotreatment of the two peptidylic inhibitors, QBP1 and L1P3V8, for their brain-targeting, and such a novel therapeutic strategy was found to be effective on a transgenic polyglutamine disease mouse model.


Subject(s)
Carrier Proteins/administration & dosage , Heredodegenerative Disorders, Nervous System/drug therapy , Oligopeptides/administration & dosage , Peptides/administration & dosage , Peptides/metabolism , RNA, Messenger/antagonists & inhibitors , Administration, Intranasal , Animals , Behavior, Animal/drug effects , Brain/metabolism , Carrier Proteins/pharmacokinetics , Disease Models, Animal , Drug Administration Schedule , Drug Evaluation, Preclinical , Drug Therapy, Combination/methods , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/pathology , Humans , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligopeptides/pharmacokinetics , Peptides/pharmacokinetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Trinucleotide Repeat Expansion/genetics
SELECTION OF CITATIONS
SEARCH DETAIL