Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 201
Filter
1.
Cell ; 164(5): 836-7, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26919420

ABSTRACT

Posttranslational modifications control microtubule behavior, yet assigning roles to particular signals was hampered by lack of defined in vitro systems. In this issue of Cell, Valenstein and Roll-Mecak establish a biochemical platform to interrogate consequences of microtubule polyglutamylation, thereby providing important insights into the specificity and quantitative nature of cellular information transfer.


Subject(s)
Adenosine Triphosphatases/metabolism , Microtubules/metabolism , Protein Processing, Post-Translational , Spastic Paraplegia, Hereditary/metabolism , Tubulin/metabolism , Humans
2.
Cell ; 164(5): 911-21, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26875866

ABSTRACT

Microtubule-severing enzymes are critical for the biogenesis and maintenance of complex microtubule arrays in axons, spindles, and cilia where tubulin detyrosination, acetylation, and glutamylation are abundant. These modifications exhibit stereotyped patterns suggesting spatial and temporal control of microtubule functions. Using human-engineered and differentially modified microtubules we find that glutamylation is the main regulator of the hereditary spastic paraplegia microtubule severing enzyme spastin. Glutamylation acts as a rheostat and tunes microtubule severing as a function of glutamate number added per tubulin. Unexpectedly, glutamylation is a non-linear biphasic tuner and becomes inhibitory beyond a threshold. Furthermore, the inhibitory effect of localized glutamylation propagates across neighboring microtubules, modulating severing in trans. Our work provides the first quantitative evidence for a graded response to a tubulin posttranslational modification and a biochemical link between tubulin glutamylation and complex architectures of microtubule arrays such as those in neurons where spastin deficiency causes disease.


Subject(s)
Adenosine Triphosphatases/metabolism , Microtubules/metabolism , Protein Processing, Post-Translational , Spastic Paraplegia, Hereditary/metabolism , Tubulin/metabolism , Biomechanical Phenomena , Glutamic Acid/metabolism , Humans , Neurons/metabolism , Protein Engineering , Spastic Paraplegia, Hereditary/pathology , Spastin
3.
J Biol Chem ; 300(5): 107259, 2024 May.
Article in English | MEDLINE | ID: mdl-38582453

ABSTRACT

Selenoprotein I (SELENOI) catalyzes the final reaction of the CDP-ethanolamine branch of the Kennedy pathway, generating the phospholipids phosphatidylethanolamine (PE) and plasmenyl-PE. Plasmenyl-PE is a key component of myelin and is characterized by a vinyl ether bond that preferentially reacts with oxidants, thus serves as a sacrificial antioxidant. In humans, multiple loss-of-function mutations in genes affecting plasmenyl-PE metabolism have been implicated in hereditary spastic paraplegia, including SELENOI. Herein, we developed a mouse model of nervous system-restricted SELENOI deficiency that circumvents embryonic lethality caused by constitutive deletion and recapitulates phenotypic features of hereditary spastic paraplegia. Resulting mice exhibited pronounced alterations in brain lipid composition, which coincided with motor deficits and neuropathology including hypomyelination, elevated reactive gliosis, and microcephaly. Further studies revealed increased lipid peroxidation in oligodendrocyte lineage cells and disrupted oligodendrocyte maturation both in vivo and in vitro. Altogether, these findings detail a critical role for SELENOI-derived plasmenyl-PE in myelination that is of paramount importance for neurodevelopment.


Subject(s)
Homeostasis , Lipid Metabolism , Myelin Sheath , Oligodendroglia , Selenoproteins , Animals , Humans , Mice , Brain/metabolism , Brain/pathology , Lipid Peroxidation , Mice, Knockout , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Phosphatidylethanolamines/metabolism , Phospholipid Ethers/metabolism , Plasmalogens/metabolism , Selenoproteins/metabolism , Selenoproteins/genetics , Spastic Paraplegia, Hereditary/metabolism , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/pathology
4.
Brain ; 147(10): 3534-3546, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-38551087

ABSTRACT

Hereditary spastic paraplegias (HSPs) are degenerative motor neuron diseases characterized by progressive spasticity and weakness in the lower limbs. The most common form of HSP is due to SPG4 gene haploinsufficiency. SPG4 encodes the microtubule severing enzyme spastin. Although, there is no cure for SPG4-HSP, strategies to induce a spastin recovery are emerging as promising therapeutic approaches. Spastin protein levels are regulated by poly-ubiquitination and proteasomal-mediated degradation, in a neddylation-dependent manner. However, the molecular players involved in this regulation are unknown. Here, we show that the Cullin-4-RING E3 ubiquitin ligase complex (CRL4) regulates spastin stability. Inhibition of CRL4 increases spastin levels by preventing its poly-ubiquitination and subsequent degradation in spastin-proficient and in patient derived SPG4 haploinsufficient cells. To evaluate the role of CRL4 complex in spastin regulation in vivo, we developed a Drosophila melanogaster model of SPG4 haploinsufficiency which show alterations of synapse morphology and locomotor activity, recapitulating phenotypical defects observed in patients. Downregulation of the CRL4 complex, highly conserved in Drosophila, rescues spastin levels and the phenotypical defects observed in flies. As a proof of concept of possible pharmacological treatments, we demonstrate a recovery of spastin levels and amelioration of the SPG4-HSP-associated defects both in the fly model and in patient-derived cells by chemical inactivation of the CRL4 complex with NSC1892. Taken together, these findings show that CRL4 contributes to spastin stability regulation and that it is possible to induce spastin recovery and rescue of SPG4-HSP defects by blocking the CRL4-mediated spastin degradation.


Subject(s)
Disease Models, Animal , Drosophila melanogaster , Spastic Paraplegia, Hereditary , Spastin , Animals , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/drug therapy , Spastic Paraplegia, Hereditary/metabolism , Spastin/metabolism , Spastin/genetics , Humans , Cullin Proteins/metabolism , Cullin Proteins/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Haploinsufficiency , Ubiquitination/drug effects
5.
Proc Natl Acad Sci U S A ; 119(40): e2210649119, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36161950

ABSTRACT

Molecular pathways that intrinsically regulate neuronal maintenance are poorly understood, but rare pathogenic mutations that underlie neurodegenerative disease can offer important insights into the mechanisms that facilitate lifelong neuronal function. Here, we leverage a rat model to demonstrate directly that the TFG p.R106C variant implicated previously in complicated forms of hereditary spastic paraplegia (HSP) underlies progressive spastic paraparesis with accompanying ventriculomegaly and thinning of the corpus callosum, consistent with disease phenotypes identified in adolescent patients. Analyses of primary cortical neurons obtained from CRISPR-Cas9-edited animals reveal a kinetic delay in biosynthetic secretory protein transport from the endoplasmic reticulum (ER), in agreement with prior induced pluripotent stem cell-based studies. Moreover, we identify an unexpected role for TFG in the trafficking of Rab4A-positive recycling endosomes specifically within axons and dendrites. Impaired TFG function compromises the transport of at least a subset of endosomal cargoes, which we show results in down-regulated inhibitory receptor signaling that may contribute to excitation-inhibition imbalances. In contrast, the morphology and trafficking of other organelles, including mitochondria and lysosomes, are unaffected by the TFG p.R106C mutation. Our findings demonstrate a multifaceted role for TFG in secretory and endosomal protein sorting that is unique to cells of the central nervous system and highlight the importance of these pathways to maintenance of corticospinal tract motor neurons.


Subject(s)
Endosomes , Motor Neurons , Protein Transport , Animals , Rats , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Motor Neurons/metabolism , Neurodegenerative Diseases/metabolism , Proteins/metabolism , Spastic Paraplegia, Hereditary/metabolism
6.
Neurobiol Dis ; 199: 106564, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38876323

ABSTRACT

Biallelic variants in the SPG11 gene account for the most common form of autosomal recessive hereditary spastic paraplegia characterized by motor and cognitive impairment, with currently no therapeutic option. We previously observed in a Spg11 knockout mouse that neurodegeneration is associated with accumulation of gangliosides in lysosomes. To test whether a substrate reduction therapy could be a therapeutic option, we downregulated the key enzyme involved in ganglioside biosynthesis using an AAV-PHP.eB viral vector expressing a miRNA targeting St3gal5. Downregulation of St3gal5 in Spg11 knockout mice prevented the accumulation of gangliosides, delayed the onset of motor and cognitive symptoms, and prevented the upregulation of serum levels of neurofilament light chain, a biomarker widely used in neurodegenerative diseases. Importantly, similar results were observed when Spg11 knockout mice were administrated venglustat, a pharmacological inhibitor of glucosylceramide synthase expected to decrease ganglioside synthesis. Downregulation of St3gal5 or venglustat administration in Spg11 knockout mice strongly decreased the formation of axonal spheroids, previously associated with impaired trafficking. Venglustat had similar effect on cultured human SPG11 neurons. In conclusion, this work identifies the first disease-modifying therapeutic strategy in SPG11, and provides data supporting its relevance for therapeutic testing in SPG11 patients.


Subject(s)
Gangliosides , Mice, Knockout , Spastic Paraplegia, Hereditary , Animals , Humans , Mice , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/genetics , Gangliosides/metabolism , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Mice, Inbred C57BL , Neurofilament Proteins , Neurons/metabolism , Proteins/genetics , Proteins/metabolism , Sialyltransferases/genetics , Sialyltransferases/deficiency , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism
7.
Neurobiol Dis ; 199: 106556, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38851544

ABSTRACT

Mutation of the ATL1 gene is one of the most common causes of hereditary spastic paraplegia (HSP), a group of genetic neurodegenerative conditions characterised by distal axonal degeneration of the corticospinal tract axons. Atlastin-1, the protein encoded by ATL1, is one of three mammalian atlastins, which are homologous dynamin-like GTPases that control endoplasmic reticulum (ER) morphology by fusing tubules to form the three-way junctions that characterise ER networks. However, it is not clear whether atlastin-1 is required for correct ER morphology in human neurons and if so what the functional consequences of lack of atlastin-1 are. Using CRISPR-inhibition we generated human cortical neurons lacking atlastin-1. We demonstrate that ER morphology was altered in these neurons, with a reduced number of three-way junctions. Neurons lacking atlastin-1 had longer endosomal tubules, suggestive of defective tubule fission. This was accompanied by reduced lysosomal proteolytic capacity. As well as demonstrating that atlastin-1 is required for correct ER morphology in human neurons, our results indicate that lack of a classical ER-shaping protein such as atlastin-1 may cause altered endosomal tubulation and lysosomal proteolytic dysfunction. Furthermore, they strengthen the idea that defective lysosome function contributes to the pathogenesis of a broad group of HSPs, including those where the primary localisation of the protein involved is not at the endolysosomal system.


Subject(s)
Cerebral Cortex , Endosomes , Lysosomes , Membrane Proteins , Neurons , Proteolysis , Humans , Lysosomes/metabolism , Neurons/metabolism , Neurons/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Endosomes/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , Endoplasmic Reticulum/metabolism , Cells, Cultured , Spastic Paraplegia, Hereditary/metabolism , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/pathology
8.
Hum Mol Genet ; 31(16): 2693-2710, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35313342

ABSTRACT

Hereditary spastic paraplegia type 15 (HSP15) is a neurodegenerative condition caused by the inability to produce SPG15 protein, which leads to lysosomal swelling. However, the link between lysosomal aberrations and neuronal death is poorly explored. To uncover the functional consequences of lysosomal aberrations in disease pathogenesis, we analyze human dermal fibroblasts from HSP15 patients as well as primary cortical neurons derived from an SPG15 knockout (KO) mouse model. We find that SPG15 protein loss induces defective anterograde transport, impaired neurite outgrowth, axonal swelling and reduced autophagic flux in association with the onset of lysosomal abnormalities. Additionally, we observe lipid accumulation within the lysosomal compartment, suggesting that distortions in cellular lipid homeostasis are intertwined with lysosomal alterations. We further demonstrate that SPG15 KO neurons exhibit synaptic dysfunction, accompanied by augmented vulnerability to glutamate-induced excitotoxicity. Overall, our study establishes an intimate link between lysosomal aberrations, lipid metabolism and electrophysiological impairments, suggesting that lysosomal defects are at the core of multiple neurodegenerative disease processes in HSP15.


Subject(s)
Neurodegenerative Diseases , Spastic Paraplegia, Hereditary , Animals , Carrier Proteins/metabolism , Humans , Lipid Metabolism/genetics , Lipids , Lysosomes/metabolism , Mice , Neurodegenerative Diseases/metabolism , Proteins/metabolism , Retinal Degeneration , Spastic Paraplegia, Hereditary/metabolism
9.
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
10.
J Biol Chem ; 298(1): 101438, 2022 01.
Article in English | MEDLINE | ID: mdl-34808209

ABSTRACT

Hereditary spastic paraplegia (HSP) comprises a heterogeneous group of neuropathies affecting upper motor neurons and causing progressive gait disorder. Mutations in the gene SPG3A/atlastin-1 (ATL1), encoding a dynamin superfamily member, which utilizes the energy from GTP hydrolysis for membrane tethering and fusion to promote the formation of a highly branched, smooth endoplasmic reticulum (ER), account for approximately 10% of all HSP cases. The continued discovery and characterization of novel disease mutations are crucial for our understanding of HSP pathogenesis and potential treatments. Here, we report a novel disease-causing, in-frame insertion in the ATL1 gene, leading to inclusion of an additional asparagine residue at position 417 (N417ins). This mutation correlates with complex, early-onset spastic quadriplegia affecting all four extremities, generalized dystonia, and a thinning of the corpus callosum. We show using limited proteolysis and FRET-based studies that this novel insertion affects a region in the protein central to intramolecular interactions and GTPase-driven conformational change, and that this insertion mutation is associated with an aberrant prehydrolysis state. While GTPase activity remains unaffected by the insertion, membrane tethering is increased, indicative of a gain-of-function disease mechanism uncommon for ATL1-associated pathologies. In conclusion, our results identify a novel insertion mutation with altered membrane tethering activity that is associated with spastic quadriplegia, potentially uncovering a broad spectrum of molecular mechanisms that may affect neuronal function.


Subject(s)
GTP-Binding Proteins , Membrane Proteins , Mutation , Spastic Paraplegia, Hereditary , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutagenesis, Insertional , Protein Conformation , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism
11.
Neurobiol Dis ; 187: 106293, 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37709208

ABSTRACT

Spastic paraplegia type 11 (SPG11) is a common autosomal recessive form of hereditary spastic paraplegia (HSP) characterized by the degeneration of cortical motor neuron axons, leading to muscle spasticity and weakness. Impaired lipid trafficking is an emerging pathology in neurodegenerative diseases including SPG11, though its role in axonal degeneration of human SPG11 neurons remains unknown. Here, we established a pluripotent stem cell-based SPG11 model by knocking down the SPG11 gene in human embryonic stem cells (hESCs). These stem cells were then differentiated into cortical projection neurons (PNs), the cell types affected in HSP patients, to examine axonal defects and cholesterol distributions. Our data revealed that SPG11 deficiency led to reduced axonal outgrowth, impaired axonal transport, and accumulated swellings, recapitulating disease-specific phenotypes. In SPG11-knockdown neurons, cholesterol was accumulated in lysosome and reduced in plasma membrane, revealing impairments in cholesterol trafficking. Strikingly, the liver-X-receptor (LXR) agonists restored cholesterol homeostasis, leading to the rescue of subsequent axonal defects in SPG11-deficient cortical PNs. To further determine the implication of impaired cholesterol homeostasis in SPG11, we examined the cholesterol distribution in cortical PNs generated from SPG11 disease-mutation knock-in hESCs, and observed a similar cholesterol trafficking impairment. Moreover, LXR agonists rescued the aberrant cholesterol distribution and mitigated the degeneration of SPG11 disease-mutated neurons. Taken together, our data demonstrate impaired cholesterol trafficking underlying axonal degeneration of SPG11 human neurons, and highlight the therapeutic potential of LXR agonists for SPG11 through restoring cholesterol homeostasis.


Subject(s)
Spastic Paraplegia, Hereditary , Humans , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , Proteins/metabolism , Neurons/metabolism , Mutation , Cholesterol/metabolism , Liver/pathology
12.
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
13.
Biochem Biophys Res Commun ; 643: 77-87, 2023 02 05.
Article in English | MEDLINE | ID: mdl-36587525

ABSTRACT

Investigating novel mechanisms of neurite outgrowth via cytoskeleton is critical for developing therapeutic strategies against neural disorders. Rab3A is a vesicle-related protein distributed throughout the nervous system, but the detailed mechanism related to cytoskeleton remains largely unknown. Our previous reports show that spastin serves microtubule to regulate neurite outgrowth. Here, we asked whether Rab3A could function via modulating spastin during neuronal development. The results revealed that Rab3A colocalized with spastin in cultured hippocampal neurons. Immunoprecipitation assays showed that Rab3A physically interacted with spastin in rat brain lysates. Rab3A overexpression significantly induced spastin degradation; this effect was reversed by leupeptin- or MG-132- administration, suggesting the lysosomal and ubiquitin-mediated degradation system. Immunofluorescence staining further confirmed that Rab3A and spastin immune-colocalized with the lysosome marker lysotracker. In COS7 cells, Rab3A overexpression significantly downregulated spastin expression and abolished the spastin-mediated microtubule severing. Furthermore, overexpression inhibited while genetic knockdown of Rab3A promoted neurite outgrowth. However, this inhibitory effect on neurite outgrowth in hippocampal neurons could be reversed via co-transfection of spastin, indicating that Rab3A functions via its interaction protein spastin. In general, our data identify an interaction between Rab3A and spastin, and this interaction affects the protein stability of spastin and eliminates its microtubule severing function, thereby modulating neurite outgrowth.


Subject(s)
Adenosine Triphosphatases , Spastic Paraplegia, Hereditary , Animals , Rats , Adenosine Triphosphatases/metabolism , Neurites/metabolism , Neuronal Outgrowth , Neurons/metabolism , rab3A GTP-Binding Protein , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , Spastin/metabolism , Spastin/pharmacology
14.
Biochem Biophys Res Commun ; 673: 169-174, 2023 09 17.
Article in English | MEDLINE | ID: mdl-37392480

ABSTRACT

Strumpellin/Wiskott-Aldrich syndrome protein and SCAR homologue (WASH) complex subunit 5 (WASHC5) is a core component of the WASH complex, and its mutations confer pathogenicity for hereditary spastic paraplegia (HSP) type SPG8, a rare neurodegenerative gait disorder. WASH complex activates actin-related protein-2/3-mediated actin polymerization and plays a pivotal role in intracellular membrane trafficking in endosomes. In this study, we examined the role of strumpellin in the regulation of structural plasticity of cortical neurons involved in gait coordination. Administration of a lentivirus containing a strumpellin-targeting short hairpin RNA (shRNA) to cortical motor neurons lead to abnormal motor coordination in mice. Strumpellin knockdown using shRNA attenuated dendritic arborization and synapse formation in cultured cortical neurons, and this effect was rescued by wild-type strumpellin expression. Compared with the wild-type, strumpellin mutants N471D or V626F identified in patients with SPG8 exhibited no differences in rescuing the defects. Moreover, the number of F-actin clusters in neuronal dendrites was decreased by strumpellin knockdown and rescued by strumpellin expression. In conclusion, our results indicate that strumpellin regulates the structural plasticity of cortical neurons via actin polymerization.


Subject(s)
Actins , Spastic Paraplegia, Hereditary , Animals , Mice , Actins/metabolism , Endosomes/metabolism , Gait , Neurons/metabolism , RNA, Small Interfering/metabolism , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism
15.
Mol Cell ; 60(1): 89-104, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26431026

ABSTRACT

Hereditary spastic paraplegias (HSPs) are a diverse group of neurodegenerative diseases that are characterized by axonopathy of the corticospinal motor neurons. A mutation in the gene encoding for Tectonin ß-propeller containing protein 2 (TECPR2) causes HSP that is complicated by neurological symptoms. While TECPR2 is a human ATG8 binding protein and positive regulator of autophagy, the exact function of TECPR2 is unknown. Here, we show that TECPR2 associates with several trafficking components, among them the COPII coat protein SEC24D. TECPR2 is required for stabilization of SEC24D protein levels, maintenance of functional ER exit sites (ERES), and efficient ER export in a manner dependent on binding to lipidated LC3C. TECPR2-deficient HSP patient cells display alterations in SEC24D abundance and ER export efficiency. Additionally, TECPR2 and LC3C are required for autophagosome formation, possibly through maintaining functional ERES. Collectively, these results reveal that TECPR2 functions as molecular scaffold linking early secretion pathway and autophagy.


Subject(s)
Autophagy , Carrier Proteins/metabolism , Endoplasmic Reticulum/metabolism , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Transport , Spastic Paraplegia, Hereditary/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins , Carrier Proteins/genetics , HeLa Cells , Humans , Mutation , Nerve Tissue Proteins/genetics , Spastic Paraplegia, Hereditary/metabolism , Vesicular Transport Proteins/metabolism
16.
Neurogenetics ; 23(3): 167-177, 2022 07.
Article in English | MEDLINE | ID: mdl-35397036

ABSTRACT

The syndromic group of hereditary spastic paraplegias has a heterogeneous clinical profile and a broad differential diagnosis, including neurometabolic disorders that are potentially treatable. This group includes 5,10-methylenetetrahydrofolate reductase deficiency, cobalamin C deficiency disease, dopamine responsive dystonia, cerebrotendinous xanthomatosis, biotinidase deficiency, GLUT1 deficiency syndrome, delta-e-pyrroline-carboxylase-synthetase deficiency, hyperonithinemia-hyperammonemia-homocitrullinuria syndrome, arginase deficiency, multiple carboxylase deficiency, and X-linked adrenoleukodystrophy. This review describes these diseases in detail, highlighting the importance of early diagnosis and effective treatment aiming at preserving functionality and quality of life in these patients. For the purpose of this study, we carried a non-systematic review on PUBMED, finding an initial sample of 122 papers; upon refining, 41 articles were found relevant to this review. Subsequently, we added review articles and works with historical relevance, totalizing 76 references. An adequate diagnostic workup in patients presenting with spastic paraplegia phenotype should include screening for these rare conditions, followed by parsimonious ancillary investigation.


Subject(s)
Homocystinuria , Spastic Paraplegia, Hereditary , Humans , Muscle Spasticity , Quality of Life , Spastic Paraplegia, Hereditary/diagnosis , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , Vitamin B 12 Deficiency/congenital
17.
Hum Mol Genet ; 29(2): 320-334, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31915823

ABSTRACT

Deficiency of the adaptor protein complex 4 (AP-4) leads to childhood-onset hereditary spastic paraplegia (AP-4-HSP): SPG47 (AP4B1), SPG50 (AP4M1), SPG51 (AP4E1) and SPG52 (AP4S1). This study aims to evaluate the impact of loss-of-function variants in AP-4 subunits on intracellular protein trafficking using patient-derived cells. We investigated 15 patient-derived fibroblast lines and generated six lines of induced pluripotent stem cell (iPSC)-derived neurons covering a wide range of AP-4 variants. All patient-derived fibroblasts showed reduced levels of the AP4E1 subunit, a surrogate for levels of the AP-4 complex. The autophagy protein ATG9A accumulated in the trans-Golgi network and was depleted from peripheral compartments. Western blot analysis demonstrated a 3-5-fold increase in ATG9A expression in patient lines. ATG9A was redistributed upon re-expression of AP4B1 arguing that mistrafficking of ATG9A is AP-4-dependent. Examining the downstream effects of ATG9A mislocalization, we found that autophagic flux was intact in patient-derived fibroblasts both under nutrient-rich conditions and when autophagy is stimulated. Mitochondrial metabolism and intracellular iron content remained unchanged. In iPSC-derived cortical neurons from patients with AP4B1-associated SPG47, AP-4 subunit levels were reduced while ATG9A accumulated in the trans-Golgi network. Levels of the autophagy marker LC3-II were reduced, suggesting a neuron-specific alteration in autophagosome turnover. Neurite outgrowth and branching were reduced in AP-4-HSP neurons pointing to a role of AP-4-mediated protein trafficking in neuronal development. Collectively, our results establish ATG9A mislocalization as a key marker of AP-4 deficiency in patient-derived cells, including the first human neuron model of AP-4-HSP, which will aid diagnostic and therapeutic studies.


Subject(s)
Adaptor Protein Complex 4/genetics , Adaptor Protein Complex 4/metabolism , Autophagy-Related Proteins/metabolism , Membrane Proteins/metabolism , Protein Transport/genetics , Spastic Paraplegia, Hereditary/metabolism , Vesicular Transport Proteins/metabolism , trans-Golgi Network/metabolism , Adaptor Protein Complex 4/deficiency , Adaptor Protein Complex beta Subunits/metabolism , Adolescent , Autophagosomes/metabolism , Autophagy/genetics , Cell Line , Child , Child, Preschool , Female , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Iron/metabolism , Loss of Function Mutation , Male , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Neurogenesis/genetics , Neurons/metabolism , Spastic Paraplegia, Hereditary/genetics , trans-Golgi Network/genetics
18.
Mol Genet Metab ; 137(4): 436-444, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34183250

ABSTRACT

Early-onset forms of hereditary spastic paraplegia and inborn errors of metabolism that present with spastic diplegia are among the most common "mimics" of cerebral palsy. Early detection of these heterogenous genetic disorders can inform genetic counseling, anticipatory guidance, and improve outcomes, particularly where specific treatments exist. The diagnosis relies on clinical pattern recognition, biochemical testing, neuroimaging, and increasingly next-generation sequencing-based molecular testing. In this short review, we summarize the clinical and molecular understanding of: 1) childhood-onset and complex forms of hereditary spastic paraplegia (SPG5, SPG7, SPG11, SPG15, SPG35, SPG47, SPG48, SPG50, SPG51, SPG52) and, 2) the most common inborn errors of metabolism that present with phenotypes that resemble hereditary spastic paraplegia.


Subject(s)
Metabolism, Inborn Errors , Retinal Degeneration , Spastic Paraplegia, Hereditary , Child , Humans , Spastic Paraplegia, Hereditary/diagnosis , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , Phenotype , High-Throughput Nucleotide Sequencing , Mutation , Proteins/genetics
19.
Neuropathol Appl Neurobiol ; 48(1): e12750, 2022 02.
Article in English | MEDLINE | ID: mdl-34312900

ABSTRACT

AIMS: We investigated N471D WASH complex subunit strumpellin (Washc5) knock-in and Washc5 knock-out mice as models for hereditary spastic paraplegia type 8 (SPG8). METHODS: We generated heterozygous and homozygous N471D Washc5 knock-in mice and subjected them to a comprehensive clinical, morphological and laboratory parameter screen, and gait analyses. Brain tissue was used for proteomic analysis. Furthermore, we generated heterozygous Washc5 knock-out mice. WASH complex subunit strumpellin expression was determined by qPCR and immunoblotting. RESULTS: Homozygous N471D Washc5 knock-in mice showed mild dilated cardiomyopathy, decreased acoustic startle reactivity, thinner eye lenses, increased alkaline phosphatase and potassium levels and increased white blood cell counts. Gait analyses revealed multiple aberrations indicative of locomotor instability. Similarly, the clinical chemistry, haematology and gait parameters of heterozygous mice also deviated from the values expected for healthy animals, albeit to a lesser extent. Proteomic analysis of brain tissue depicted consistent upregulation of BPTF and downregulation of KLHL11 in heterozygous and homozygous knock-in mice. WASHC5-related protein interaction partners and complexes showed no change in abundancies. Heterozygous Washc5 knock-out mice showing normal WASHC5 levels could not be bred to homozygosity. CONCLUSIONS: While biallelic ablation of Washc5 was prenatally lethal, expression of N471D mutated WASHC5 led to several mild clinical and laboratory parameter abnormalities, but not to a typical SPG8 phenotype. The consistent upregulation of BPTF and downregulation of KLHL11 suggest mechanistic links between the expression of N471D mutated WASHC5 and the roles of both proteins in neurodegeneration and protein quality control, respectively.


Subject(s)
Proteomics , Spastic Paraplegia, Hereditary , Animals , Brain/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Mutation , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism
20.
Int J Mol Sci ; 23(8)2022 Apr 14.
Article in English | MEDLINE | ID: mdl-35457156

ABSTRACT

Adhesion molecules regulate cell proliferation, migration, survival, neuritogenesis, synapse formation and synaptic plasticity during the nervous system's development and in the adult. Among such molecules, the neural cell adhesion molecule L1 contributes to these functions during development, and in synapse formation, synaptic plasticity and regeneration after trauma. Proteolytic cleavage of L1 by different proteases is essential for these functions. A proteolytic fragment of 70 kDa (abbreviated L1-70) comprising part of the extracellular domain and the transmembrane and intracellular domains was shown to interact with mitochondrial proteins and is suggested to be involved in mitochondrial functions. To further determine the role of L1-70 in mitochondria, we generated two lines of gene-edited mice expressing full-length L1, but no or only low levels of L1-70. We showed that in the absence of L1-70, mitochondria in cultured cerebellar neurons move more retrogradely and exhibit reduced mitochondrial membrane potential, impaired Complex I activity and lower ATP levels compared to wild-type littermates. Neither neuronal migration, neuronal survival nor neuritogenesis in these mutants were stimulated with a function-triggering L1 antibody or with small agonistic L1 mimetics. These results suggest that L1-70 is important for mitochondrial homeostasis and that its absence contributes to the L1 syndrome phenotypes.


Subject(s)
Neural Cell Adhesion Molecule L1 , Spastic Paraplegia, Hereditary , Animals , Mice , Mitochondria/genetics , Mitochondria/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neurites/metabolism , Neurogenesis/genetics , Neurons/metabolism , Spastic Paraplegia, Hereditary/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL