Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters

Publication year range
1.
Am J Hum Genet ; 108(10): 2017-2023, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34587489

ABSTRACT

ABHD16A (abhydrolase domain-containing protein 16A, phospholipase) encodes the major phosphatidylserine (PS) lipase in the brain. PS lipase synthesizes lysophosphatidylserine, an important signaling lipid that functions in the mammalian central nervous system. ABHD16A has not yet been associated with a human disease. In this report, we present a cohort of 11 affected individuals from six unrelated families with a complicated form of hereditary spastic paraplegia (HSP) who carry bi-allelic deleterious variants in ABHD16A. Affected individuals present with a similar phenotype consisting of global developmental delay/intellectual disability, progressive spasticity affecting the upper and lower limbs, and corpus callosum and white matter anomalies. Immunoblot analysis on extracts from fibroblasts from four affected individuals demonstrated little to no ABHD16A protein levels compared to controls. Our findings add ABHD16A to the growing list of lipid genes in which dysregulation can cause complicated forms of HSP and begin to describe the molecular etiology of this condition.


Subject(s)
Cerebral Palsy/pathology , Intellectual Disability/pathology , Leukoencephalopathies/pathology , Monoacylglycerol Lipases/genetics , Mutation , Spastic Paraplegia, Hereditary/pathology , Adolescent , Adult , Cerebral Palsy/etiology , Cerebral Palsy/metabolism , Child , Child, Preschool , Cohort Studies , Female , Humans , Intellectual Disability/etiology , Intellectual Disability/metabolism , Leukoencephalopathies/etiology , Leukoencephalopathies/metabolism , Male , Monoacylglycerol Lipases/deficiency , Pedigree , Phenotype , Spastic Paraplegia, Hereditary/etiology , Spastic Paraplegia, Hereditary/metabolism , Young Adult
2.
J Clin Lab Anal ; 35(6): e23772, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33826176

ABSTRACT

BACKGROUND: Hereditary spastic paraplegia is a rare familial hereditary neurodegenerative disease caused by multiple autosomal dominant mutations. More than 50 mutant genes have been reported to be associated with this disease. METHODS: In this study, we have reported a rare insertion mutation site in PRRT2 that caused a familial disorder of hereditary spastic paraplegia accompanied by polyneuropathy. RESULTS: We used second-generation sequencing of samples of the proband's familial genome and found an insertion mutation of C/CC in NM_001256443:c.641dupC that was localized to the second exon of PRRT2. This functional mutation can cause an amino acid sequence change (arginine >proline) and dysfunctional neuronal transmembrane proteins, which might have been related to the onset of hereditary spastic paraplegia accompanied by polyneuropathy in the family reported in this study. CONCLUSION: The discovery of this mutation site provides an important theoretical basis for specific gene-based diagnosis and treatment of hereditary spastic paraplegia.


Subject(s)
INDEL Mutation , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Polyneuropathies/genetics , Spastic Paraplegia, Hereditary/genetics , Female , Humans , Male , Middle Aged , Pedigree , Polyneuropathies/etiology , Spastic Paraplegia, Hereditary/etiology
3.
J Inherit Metab Dis ; 43(1): 51-62, 2020 01.
Article in English | MEDLINE | ID: mdl-30854657

ABSTRACT

Autophagy is a fundamental and conserved catabolic pathway that mediates the degradation of macromolecules and organelles in lysosomes. Autophagy is particularly important to postmitotic and metabolically active cells such as neurons. The complex architecture of neurons and their long axons pose additional challenges for efficient recycling of cargo. Not surprisingly autophagy is required for normal central nervous system development and function. Several single-gene disorders of the autophagy pathway have been discovered in recent years giving rise to a novel group of inborn errors of metabolism referred to as congenital disorders of autophagy. While these disorders are heterogeneous, they share several clinical and molecular characteristics including a prominent and progressive involvement of the central nervous system leading to brain malformations, developmental delay, intellectual disability, epilepsy, movement disorders, and cognitive decline. On brain magnetic resonance imaging a predominant involvement of the corpus callosum, the corticospinal tracts and the cerebellum are noted. A storage disease phenotype is present in some diseases, underscoring both clinical and molecular overlaps to lysosomal storage diseases. This review provides an update on the clinical, imaging, and genetic spectrum of congenital disorders of autophagy and highlights the importance of this pathway for neurometabolism and childhood-onset neurological diseases.


Subject(s)
Autophagy/physiology , Developmental Disabilities/etiology , Neurodegenerative Diseases/etiology , Agenesis of Corpus Callosum/etiology , Agenesis of Corpus Callosum/genetics , Brain/pathology , Cataract/etiology , Cataract/genetics , Child , Developmental Disabilities/genetics , Humans , Magnetic Resonance Imaging , Neurodegenerative Diseases/genetics , Proteins/genetics , Spastic Paraplegia, Hereditary/etiology , Spastic Paraplegia, Hereditary/genetics
4.
Mov Disord ; 26(3): 553-6, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21381113

ABSTRACT

BACKGROUND: Autosomal recessive hereditary spastic paraplegia with thin corpus callosum is a neurodegenerative disorder characterized by spastic paraparesis, cognitive impairment, and peripheral neuropathy. The neuroradiologic hallmarks are thin corpus callosum and periventricular white matter changes. Mutations in the SPG11 gene have been identified to be a major cause of autosomal recessive hereditary spastic paraplegia with thin corpus callosum and recently also proven to be responsible for juvenile parkinsonism associated with spastic paraplegia. METHODS: We describe one Italian autosomal recessive hereditary spastic paraplegia with thin corpus callosum patient who unusually presented at onset, 16 years, with parkinsonism-like features, responsive to dopaminergic therapy. Then the clinical picture evolved and became more complex. A brain magnetic resonance imaging scan showed thin corpus callosum and hyperintense T(2)-weighted lesions in periventricular regions, and the (123)I-ioflupane single-photon emission coupled tomography was abnormal. RESULTS: Genetic analysis detected two novel mutations, a c.3664insT variant in compound heterozygosity with a c.6331insG mutation, in SPG11. DISCUSSION: This case confirms the high genetic and clinical heterogeneity associated with SPG11 mutations. It also offers further evidence that parkinsonism may initiate autosomal recessive hereditary spastic paraplegia with thin corpus callosum and that parkinsonian symptoms can have variable dopaminergic response in these patients.


Subject(s)
Mutation/genetics , Parkinson Disease/complications , Proteins/genetics , Spastic Paraplegia, Hereditary/etiology , Spastic Paraplegia, Hereditary/genetics , Adult , Antiparkinson Agents/therapeutic use , Female , Genome-Wide Association Study/methods , Humans , Levodopa/therapeutic use , Magnetic Resonance Imaging/methods , Parkinson Disease/drug therapy , Parkinson Disease/genetics
5.
Sci Rep ; 11(1): 22248, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34782662

ABSTRACT

The present study aimed to characterize clinical and molecular data of a large cohort of subjects with childhood-onset hereditary spastic paraplegias (HSPs). A multicenter historical cohort was performed at five centers in Brazil, in which probands and affected relatives' data from consecutive families with childhood-onset HSP (onset < 12Ā years-old) were reviewed from 2011 to 2020. One hundred and six individuals (83 families) with suspicion of childhood-onset HSP were evaluated, being 68 (50 families) with solved genetic diagnosis, 6 (5 families) with candidate variants in HSP-related genes and 32 (28 families) with unsolved genetic diagnosis. The most common childhood-onset subtype was SPG4, 11/50 (22%) families with solved genetic diagnosis; followed by SPG3A, 8/50 (16%). Missense pathogenic variants in SPAST were found in 54.5% of probands, favoring the association of this type of variant to childhood-onset SPG4. Survival curves to major handicap and cross-sectional Spastic Paraplegia Rating Scale progressions confirmed the slow neurological deterioration in SPG4 and SPG3A. Most common complicating features and twenty variants not previously described in HSP-related genes were reported. These results are fundamental to understand the molecular and clinical epidemiology of childhood-onset HSP, which might help on differential diagnosis, patient care and guiding future collaborative trials for these rare diseases.


Subject(s)
Spastic Paraplegia, Hereditary/diagnosis , Spastic Paraplegia, Hereditary/etiology , Adolescent , Adult , Age of Onset , Alleles , Brazil/epidemiology , Child , Cohort Studies , Disease Management , Disease Susceptibility , Female , Genetic Predisposition to Disease , Genotype , High-Throughput Nucleotide Sequencing , Humans , Magnetic Resonance Imaging , Male , Phenotype , Population Surveillance , Spastic Paraplegia, Hereditary/epidemiology , Spastin/genetics , Symptom Assessment , Young Adult
6.
Mol Biol Cell ; 18(5): 1683-92, 2007 May.
Article in English | MEDLINE | ID: mdl-17332501

ABSTRACT

Troyer syndrome is an autosomal recessive hereditary spastic paraplegia caused by mutation in the spartin (SPG20) gene, which encodes a widely expressed protein of unknown function. This mutation results in premature protein truncation and thus might signify a loss-of-function disease mechanism. In this study, we have found that spartin is mono-ubiquitinated and functions in degradation of the epidermal growth factor receptor (EGFR). Upon EGF stimulation, spartin translocates from the cytoplasm to the plasma membrane and colocalizes with internalized EGF-Alexa. Knockdown of spartin by small interfering RNA decreases the rate of EGFR degradation and also affects EGFR internalization, recycling, or both. Furthermore, overexpression of spartin results in a prominent decrease in EGFR degradation. Taken together, our data suggest that spartin is involved in the intracellular trafficking of EGFR and that impaired endocytosis may underlie the pathogenesis of Troyer syndrome.


Subject(s)
ErbB Receptors/metabolism , Proteins/metabolism , ATPases Associated with Diverse Cellular Activities , Adaptor Proteins, Signal Transducing , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins , Cell Line , DNA, Complementary/genetics , Endocytosis , Endosomal Sorting Complexes Required for Transport , Endosomes/metabolism , Gene Expression , HeLa Cells , Humans , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mutation , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Transport , Proteins/antagonists & inhibitors , Proteins/chemistry , Proteins/genetics , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spastic Paraplegia, Hereditary/etiology , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , Syndrome , Ubiquitin/metabolism , Vacuolar Proton-Translocating ATPases , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
7.
Biol Open ; 9(9)2020 09 29.
Article in English | MEDLINE | ID: mdl-32878877

ABSTRACT

Hereditary spastic paraplegias (HSPs) are genetic neurodegenerative diseases. HSPs are characterized by lower-extremity weakness and spasticity. However, there is no specific clinical treatment strategy to prevent or reverse nerve degeneration in HSPs. Mutations in receptor expression-enhancing protein 1 (REEP1) are well-recognized and relatively common causes of autosomal dominant HSPs. REEP1 modifies the endoplasmic reticulum (ER) shape, and is implicated in the ER stress response. Defects in the ER stress response seem to be crucial mechanisms underlying HSP neurodegeneration. Here, we report that REEP1-/- mice exhibit progressive motor deficits, along with denervation of neuromuscular junctions and increased ER stress. Moreover, marked axonal degeneration and morphological abnormalities are observed. In this study, we treated both REEP1-/- and wild-type (WT) mice with salubrinal, which is a specific inhibitor of ER stress, and we observed increased nerve-muscle connections and enhanced motor functions. Our data highlight the importance of ER homeostasis in HSPs, providing new opportunities for HSP treatment.


Subject(s)
Disease Susceptibility , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Membrane Proteins/deficiency , Spastic Paraplegia, Hereditary/etiology , Spastic Paraplegia, Hereditary/metabolism , Animals , Disease Models, Animal , Disease Progression , Genetic Predisposition to Disease , Homozygote , Mice , Motor Activity , Motor Neurons/metabolism , Motor Neurons/pathology , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Spastic Paraplegia, Hereditary/diagnosis , Spastic Paraplegia, Hereditary/physiopathology
9.
Adv Exp Med Biol ; 652: 263-96, 2009.
Article in English | MEDLINE | ID: mdl-20225032

ABSTRACT

Inherited ataxias and hereditary spastic paraplegias are two heterogeneous groups of neurodegenerative disorders with a wide spectrum of clinical symptoms and also, with a remarkable number of involved loci/genes. Inherited ataxias are clinically characterized by progressive degeneration of cerebellum and spinocerebellar tracts of the spinal cord associated with a variable combination of signs of central and peripheral nervous system. Hereditary spastic paraplegias (HSPs) are characterized by slowly progressive spasticity and weakness of lower limbs, due to pyramidal tract dysfunction. The classification of these diseases is extremely difficult because of overlapping symptoms among different clinical forms. For this reason, the genetic classification for both inherited ataxias and HSP forms, based on the causative loci/genes has reached general acceptance. The aim of this review is to summarize the genetics and the pathogenic mechanisms involved in these two groups of neurodegenerative spinocerebellar disorders.


Subject(s)
Ataxia/etiology , Ataxia/genetics , Spastic Paraplegia, Hereditary/etiology , Spastic Paraplegia, Hereditary/genetics , Humans , Spastic Paraplegia, Hereditary/classification
10.
Cytoskeleton (Hoboken) ; 76(4): 289-297, 2019 04.
Article in English | MEDLINE | ID: mdl-31108029

ABSTRACT

Mutations of the SPAST gene are the chief cause of hereditary spastic paraplegia. Controversy exists in the medical community as to whether the etiology of the disease is haploinsufficiency or toxic gain-of-function properties of the mutant spastin proteins. In recognition of strong reasons that support each possible mechanism, here we present a novel perspective, based in part on new studies with mouse models and in part on the largest study to date on patients with the disease. We posit that haploinsufficiency does not cause the disease but makes the corticospinal tracts vulnerable to a second hit, which is usually the mutant spastin proteins but could also be proteins generated by mutations of other genes that may or may not cause the disease on their own.


Subject(s)
Spastic Paraplegia, Hereditary/etiology , Female , Humans , Male
11.
Sci Rep ; 9(1): 9615, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31270336

ABSTRACT

Axonal degeneration is a key pathology of neurodegenerative diseases, including hereditary spastic paraplegia (HSP), a disorder characterized by spasticity in the lower limbs. Treatments for HSP and other neurodegenerative diseases are mainly symptomatic. While iPSC-derived neurons are valuable for drug discovery and target identification, these applications require robust differentiation paradigms and rapid phenotypic read-outs ranging between hours and a few days. Using spastic paraplegia type 4 (SPG4, the most frequent HSP subtype) as an exemplar, we here present three rapid phenotypic assays for uncovering neuronal process pathologies in iPSC-derived glutamatergic cortical neurons. Specifically, these assays detected a 51% reduction in neurite outgrowth and a 60% increase in growth cone area already 24 hours after plating; axonal swellings, a hallmark of HSP pathology, was discernible after only 5 days. Remarkably, the identified phenotypes were neuron subtype-specific and not detectable in SPG4-derived GABAergic forebrain neurons. We transferred all three phenotypic assays to a 96-well setup, applied small molecules and found that a liver X receptor (LXR) agonist rescued all three phenotypes in HSP neurons, providing a potential drug target for HSP treatment. We expect this multiparametric and rapid phenotyping approach to accelerate development of therapeutic compounds for HSP and other neurodegenerative diseases.


Subject(s)
Biomarkers , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Neurons/drug effects , Neurons/metabolism , Cell Differentiation , Cells, Cultured , Haploinsufficiency , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neuronal Outgrowth , Phenotype , Spastic Paraplegia, Hereditary/drug therapy , Spastic Paraplegia, Hereditary/etiology , Spastic Paraplegia, Hereditary/metabolism , Spastin/genetics
12.
J Neurol Sci ; 210(1-2): 35-9, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12736085

ABSTRACT

A total of eight loci for autosomal dominant hereditary spastic paraplegia (ADHSP) has been mapped to chromosome 14q, 2p, 15q, 8q, 10q, 12q, 19q, 2q, respectively, among which the SPG4 gene on chromosome 2p21-22 encoding spastin, an ATPase of the AAA family, accounts for 40-50% of all ADHSP families and is expressed in both adult and fetal tissues. In this work, we reveal a novel insertion mutation in exon 11 of the SPG4 gene found in a big Chinese family composed of 47 members, including 20 affected ones, using linkage analysis. The mutation was well demonstrated to be the cause of loss of production of the functional protein by pre-termination of translation in AAA cassette region. To our knowledge, this is the first report of spastin mutation in China.


Subject(s)
Asian People/genetics , Calcium-Binding Proteins/genetics , Mutagenesis, Insertional , Spastic Paraplegia, Hereditary/genetics , Adenosine Triphosphatases , DNA Mutational Analysis/methods , Exons , Family Health , Female , Genes, Dominant , Genetic Linkage , Genotype , Humans , Male , Molecular Sequence Data , Phenotype , Restriction Mapping , Spastic Paraplegia, Hereditary/etiology , Spastin
13.
J Child Neurol ; 18(2): 140-1, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12693783

ABSTRACT

We report two siblings with purine nucleoside phosphorylase deficiency revealed by isolated spastic paraplegia, whereas symptoms of immune deficiency did not become apparent until 3 years of age. As the concurrence of immunodeficiency and neurologic problems strongly suggests the diagnosis of purine nucleoside phosphorylase deficiency, special attention should be paid to counts of lymphocytes in any infant with spastic paraplegia.


Subject(s)
Purine-Nucleoside Phosphorylase/deficiency , Spastic Paraplegia, Hereditary/etiology , Cerebral Cortex/pathology , Child, Preschool , Female , Humans , Lymphocyte Count , Magnetic Resonance Imaging , Purine-Nucleoside Phosphorylase/genetics , Siblings , Spastic Paraplegia, Hereditary/immunology , Spastic Paraplegia, Hereditary/pathology
14.
Article in Russian | MEDLINE | ID: mdl-1661531

ABSTRACT

The authors describe a family living in the Dagestan where three relative sibs, girls, suffered from familial spastic paraplegia. The variety described is marked by early debut, pronounced intrafamilial polymorphism of the disease course, autosomal recessive type of inheritance which, according to the reported data, is marked by frequently occurring combination with damage to other organs and systems (the familial spastic paraplegia "plus"). The "pure" disease variety seen in autosomal recessive type of inheritance, detection of the disease symptoms since the birth indicate that the case in question is a rare clinical variety of familial spastic paraplegia.


Subject(s)
Chromosome Aberrations/diagnosis , Spastic Paraplegia, Hereditary/diagnosis , Child , Child, Preschool , Chromosome Aberrations/etiology , Chromosome Aberrations/genetics , Chromosome Disorders , Female , Genes, Recessive/genetics , Humans , Muscle Spasticity/diagnosis , Muscle Spasticity/etiology , Muscle Spasticity/genetics , Pedigree , Spastic Paraplegia, Hereditary/etiology , Spastic Paraplegia, Hereditary/genetics
15.
J Neurol ; 261(4): 747-51, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24519355

ABSTRACT

Pathogenic mutations in CYP7B1 account for SPG5, an autosomal recessive hereditary spastic paraplegia characterized by a complex phenotype including visual problems and cerebellar dysfunction. Sensory ataxia is not usually regarded as a typical clinical feature of SPG5. The purpose of this study was to describe six patients showing features of sensory ataxia as the prominent and/or initial symptoms of SPG5. Six patients from three distinct pedigrees (three women, three men; age 49.5 Ā± 18.2 years), all presenting gait unsteadiness and frequent falls since childhood, underwent clinical and molecular investigations. All showed marked sensory ataxic gait with positive Romberg's sign, as well as severely impaired position and vibration sense. Comparatively minor signs of pyramidal involvement were also detected. In four of the patients, brain MRI showed white matter hyperintensities on T2-weighted images. An already reported homozygous c.889A>G (p.T297A) mutation in SPG5/CYP7B1 was found in five patients from two families, whereas the remaining case harbored the novel c.250_251delC/p.L84Ffs*6 and c.266A>C/p.Y89S variants. Marked and enduring sensory ataxia can be a pivotal sign in SPG5, and expands the phenotypic spectrum associated with mutations in CYP7B1.


Subject(s)
Ataxia/etiology , Ataxia/genetics , Spastic Paraplegia, Hereditary/etiology , Spastic Paraplegia, Hereditary/genetics , Steroid Hydroxylases/genetics , Adolescent , Adult , Age of Onset , Aged , Cytochrome P450 Family 7 , Exons/genetics , Female , Humans , Male , Middle Aged , Mutation/genetics , Pedigree
16.
JAMA Neurol ; 71(7): 901-4, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24797679

ABSTRACT

IMPORTANCE: Hereditary spastic paraplegia is a highly heterogeneous group of neurogenetic disorders with pure and complicated clinical phenotypes. No treatment is available for these disorders. We identified 2 unrelated families, each with 2 siblings with severe methylenetetrahydrofolate reductase (MTHFR) deficiency manifesting a complicated form of adult-onset hereditary spastic paraparesis partially responsive to betaine therapy. OBSERVATIONS: Both pairs of siblings presented with a similar combination of progressive spastic paraparesis and polyneuropathy, variably associated with behavioral changes, cognitive impairment, psychosis, seizures, and leukoencephalopathy, beginning between the ages of 29 and 50 years. By the time of diagnosis a decade later, 3 patients were ambulatory and 1 was bedridden. Investigations have revealed severe hyperhomocysteinemia and hypomethioninemia, reduced fibroblast MTHFR enzymatic activity (18%-52% of control participants), and 3 novel pathogenic MTHFR mutations, 2 as compound heterozygotes in one family and 1 as a homozygous mutation in the other family. Treatment with betaine produced a rapid decline of homocysteine by 50% to 70% in all 4 patients and, over 9 to 15 years, improved the conditions of the 3 ambulatory patients. CONCLUSIONS AND RELEVANCE: Although severe MTHFR deficiency is a rare cause of complicated spastic paraparesis in adults, it should be considered in select patients because of the potential therapeutic benefit of betaine supplementation.


Subject(s)
Betaine/pharmacology , Homocystinuria/genetics , Lipotropic Agents/pharmacology , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Muscle Spasticity/genetics , Severity of Illness Index , Spastic Paraplegia, Hereditary/etiology , Spastic Paraplegia, Hereditary/genetics , Adult , Age of Onset , Aged , Female , Homocystinuria/classification , Humans , Magnetic Resonance Imaging , Male , Methylenetetrahydrofolate Reductase (NADPH2)/classification , Methylenetetrahydrofolate Reductase (NADPH2)/drug effects , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Middle Aged , Muscle Spasticity/classification , Prospective Studies , Psychotic Disorders/classification , Psychotic Disorders/genetics , Spastic Paraplegia, Hereditary/drug therapy , Treatment Outcome
17.
Neuroscientist ; 19(4): 336-44, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22785106

ABSTRACT

Molecular protein motors play key roles in processes such as intracellular cargo transport and brain wiring, and failure of function can give rise to serious diseases. Kinesin-1, a member of the kinesin superfamily (also known as KIFs) is a two-headed motor protein that uses energy derived from ATP hydrolysis to transport diverse types of intracellular cargo toward the plus-ends of microtubules within axons. Recent studies at the level of a single molecule have provided extensive knowledge on how kinesin-1 moves along microtubules. Further elucidation of kinesin-1 movement may shed light on its influence on axon generation, thereby leading to therapies for diseases such as spastic paraplegia type 10 (SPG10), the subject of this review. SPG10 is an autosomal dominant form of hereditary spastic paraplegia caused by mutations in KIF5A, which encodes one of the isoforms of kinesin-1 (KIF5A, KIF5B, and KIF5C). Although little is known about the cargo of KIF5A, a recent study revealed an axonal transport defect of mitochondria in a KIF5A (-/-) mouse model. This review discusses the consensus moving model of kinesin-1 and the pathogenicity of SPG10 caused by defective KIF5A function.


Subject(s)
Kinesins/genetics , Spastic Paraplegia, Hereditary/genetics , Animals , Humans , Kinesins/metabolism , Mice , Mice, Knockout , Microtubules/metabolism , Spastic Paraplegia, Hereditary/etiology
19.
J Neurol Sci ; 306(1-2): 62-5, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21546041

ABSTRACT

BACKGROUND: Hereditary spastic paraplegia (HSP) is characterised in its pure form by slowly progressive spastic paraparesis. Around 40% of autosomal dominant (AD) cases are caused by mutations in SPAST, encoding spastin. PATIENTS AND METHODS: The clinical and investigation details of all patients with a SPAST mutation identified through our centre were reviewed. All published reports of SPAST mutations where the sex of patients was given were subsequently analysed in order to determine whether there is evidence of one sex being preferentially affected. RESULTS: In total 22 probable pathogenic changes were detected, including 11 novel ones. One patient carried two adjacent missense mutations. The pathogenicity of a further novel missense mutation is uncertain. Most patients had a pure phenotype, although mild peripheral neuropathy was sometimes present. The total number of patients with SPAST mutations was 27, as three of the previously known mutations were present in more than one person. The excess of males over females in our population (17:10) prompted us to review all published studies where the sex of the patients was given (n=31). A significant excess of males was identified (ratio 1.29, p=0.0007). CONCLUSIONS: Our results are consistent with data suggesting that SPAST mutations mostly cause a pure HSP phenotype. The excess of males in our sample and in published reports suggests that penetrance or severity may be sex-dependent, and merits further investigation as it may have important implications for counselling.


Subject(s)
Adenosine Triphosphatases/genetics , Genetic Predisposition to Disease/genetics , Mutation/genetics , Spastic Paraplegia, Hereditary/etiology , Spastic Paraplegia, Hereditary/genetics , Databases, Factual/statistics & numerical data , Exons/genetics , Female , Genome-Wide Association Study/methods , Humans , Male , Spastin
20.
J Clin Invest ; 120(4): 1097-110, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20200447

ABSTRACT

Hereditary spastic paraplegias (HSPs; SPG1-45) are inherited neurological disorders characterized by lower extremity spastic weakness. More than half of HSP cases result from autosomal dominant mutations in atlastin-1 (also known as SPG3A), receptor expression enhancing protein 1 (REEP1; SPG31), or spastin (SPG4). The atlastin-1 GTPase interacts with spastin, a microtubule-severing ATPase, as well as with the DP1/Yop1p and reticulon families of ER-shaping proteins, and SPG3A caused by atlastin-1 mutations has been linked pathogenically to abnormal ER morphology. Here we investigated SPG31 by analyzing the distribution, interactions, and functions of REEP1. We determined that REEP1 is structurally related to the DP1/Yop1p family of ER-shaping proteins and localizes to the ER in cultured rat cerebral cortical neurons, where it colocalizes with spastin and atlastin-1. Upon overexpression in COS7 cells, REEP1 formed protein complexes with atlastin-1 and spastin within the tubular ER, and these interactions required hydrophobic hairpin domains in each of these proteins. REEP proteins were required for ER network formation in vitro, and REEP1 also bound microtubules and promoted ER alignment along the microtubule cytoskeleton in COS7 cells. A SPG31 mutant REEP1 lacking the C-terminal cytoplasmic region did not interact with microtubules and disrupted the ER network. These data indicate that the HSP proteins atlastin-1, spastin, and REEP1 interact within the tubularER membrane in corticospinal neurons to coordinate ER shaping and microtubule dynamics. Thus, defects in tubular ER shaping and network interactions with the microtubule cytoskeleton seem to be the predominant pathogenic mechanism of HSP.


Subject(s)
Adenosine Triphosphatases/physiology , Endoplasmic Reticulum/physiology , GTP Phosphohydrolases/physiology , Membrane Transport Proteins/physiology , Microtubules/physiology , Spastic Paraplegia, Hereditary/etiology , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , GTP-Binding Proteins , Humans , Male , Membrane Proteins/physiology , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Spastin
SELECTION OF CITATIONS
SEARCH DETAIL