Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
3.
Stem Cell Res ; 66: 102998, 2023 02.
Article in English | MEDLINE | ID: mdl-36528014

ABSTRACT

The most common genetic cause of Amyotrophic Lateral Sclerosis (ALS) is the expansion of a G4C2 hexanucleotide repeat in the C9orf72 gene. The size of the repeat expansion is highly variable and a cut-off of 30 repeats has been suggested as the lower pathological limit. Repeat size variability has been observed intergenerationally and intraindividually in tissues from different organs and within the same tissue, suggesting instability of the pathological repeat expansion. In order to study this genomic instability, we established iPSCs from five members of the same family of which four carried a C9orf72 repeat expansion and one was wild-type.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Induced Pluripotent Stem Cells , Humans , Proteins/genetics , C9orf72 Protein/genetics , Induced Pluripotent Stem Cells/pathology , DNA Repeat Expansion/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Frontotemporal Dementia/genetics
4.
Article in English | MEDLINE | ID: mdl-33602014

ABSTRACT

Primary lateral sclerosis (PLS) is a rare neurodegenerative disease characterized by progressive degeneration of upper motor neurons (UMNs). Recent studies shed new light onto the cellular events that are particularly important for UMN maintenance including intracellular trafficking, mitochondrial energy homeostasis and lipid metabolism. This review summarizes these advances including the role of Alsin as a gene linked to atypical forms of juvenile PLS, and discusses wider aspects of cellular pathology that have been observed in adult forms of PLS. The review further discusses the prospects of new transgenic upper motor neuron reporter mice, human stem cell-derived UMN cultures, cerebral organoids and non-human primates as future model systems to better understand and ultimately treat PLS.


Subject(s)
Amyotrophic Lateral Sclerosis , Motor Neuron Disease , Neurodegenerative Diseases , Amyotrophic Lateral Sclerosis/genetics , Animals , Guanine Nucleotide Exchange Factors , Mice , Motor Neuron Disease/genetics , Motor Neurons
7.
Proc Natl Acad Sci U S A ; 114(12): E2486-E2493, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28270618

ABSTRACT

Numerous neurotrophic factors promote the survival of developing motor neurons but their combinatorial actions remain poorly understood; to address this, we here screened 66 combinations of 12 neurotrophic factors on pure, highly viable, and standardized embryonic mouse motor neurons isolated by a unique FACS technique. We demonstrate potent, strictly additive, survival effects of hepatocyte growth factor (HGF), ciliary neurotrophic factor (CNTF), and Artemin through specific activation of their receptor complexes in distinct subsets of lumbar motor neurons: HGF supports hindlimb motor neurons through c-Met; CNTF supports subsets of axial motor neurons through CNTFRα; and Artemin acts as the first survival factor for parasympathetic preganglionic motor neurons through GFRα3/Syndecan-3 activation. These data show that neurotrophic factors can selectively promote the survival of distinct classes of embryonic motor neurons. Similar studies on postnatal motor neurons may provide a conceptual framework for the combined therapeutic use of neurotrophic factors in degenerative motor neuron diseases such as amyotrophic lateral sclerosis, spinal muscular atrophy, and spinobulbar muscular atrophy.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Ciliary Neurotrophic Factor/metabolism , Hepatocyte Growth Factor/metabolism , Motor Neurons/metabolism , Nerve Tissue Proteins/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Cell Survival , Ciliary Neurotrophic Factor Receptor alpha Subunit/genetics , Ciliary Neurotrophic Factor Receptor alpha Subunit/metabolism , Female , Flow Cytometry , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/cytology , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Syndecan-3/genetics , Syndecan-3/metabolism
8.
Brain Pathol ; 27(4): 459-471, 2017 07.
Article in English | MEDLINE | ID: mdl-27488538

ABSTRACT

Motor neuron diseases such as amyotrophic lateral sclerosis (ALS) are now recognized as multi-system disorders also involving various non-motor neuronal cell types. The precise extent and mechanistic basis of non-motor neuron damage in human ALS and ALS animal models remain however unclear. To address this, we here studied progressive motor neuronopathy (pmn) mice carrying a missense loss-of-function mutation in tubulin binding cofactor E (TBCE). These mice manifest a particularly aggressive form of motor axon dying back and display a microtubule loss, similar to that induced by human ALS-linked TUBA4A mutations. Using whole nerve confocal imaging of pmn × thy1.2-YFP16 fluorescent reporter mice and electron microscopy, we demonstrate axonal discontinuities, bead-like spheroids and ovoids in pmn suralis nerves indicating prominent sensory neuropathy. The axonal alterations qualitatively resemble those in phrenic motor nerves but do not culminate in the loss of myelinated fibers. We further show that the pmn mutation decreases the level of TBCE, impedes microtubule polymerization in dorsal root ganglion (DRG) neurons and causes progressive loss of microtubules in large and small caliber suralis axons. Live imaging of axonal transport using GFP-tagged tetanus toxin C-fragment (GFP-TTC) demonstrates defects in microtubule-based transport in pmn DRG neurons, providing a potential explanation for the axonal alterations in sensory nerves. This study unravels sensory neuropathy as a pathological feature of mouse pmn, and discusses the potential contribution of cytoskeletal defects to sensory neuropathy in human motor neuron disease.


Subject(s)
Axonal Transport/genetics , Microtubules/metabolism , Motor Neuron Disease/complications , Peripheral Nervous System Diseases/etiology , Peripheral Nervous System Diseases/pathology , Sural Nerve/pathology , Animals , Axons/metabolism , Axons/pathology , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian , Ganglia, Spinal/cytology , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Mice, Transgenic , Microtubules/genetics , Microtubules/ultrastructure , Molecular Chaperones/genetics , Motor Neuron Disease/genetics , Motor Neuron Disease/pathology , Mutation, Missense/genetics , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Phrenic Nerve/pathology , Phrenic Nerve/ultrastructure , Polymerization , Sural Nerve/metabolism , Sural Nerve/ultrastructure
9.
Am J Hum Genet ; 99(4): 974-983, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27666369

ABSTRACT

Tubulinopathies constitute a family of neurodevelopmental/neurodegenerative disorders caused by mutations in several genes encoding tubulin isoforms. Loss-of-function mutations in TBCE, encoding one of the five tubulin-specific chaperones involved in tubulin folding and polymerization, cause two rare neurodevelopmental syndromes, hypoparathyroidism-retardation-dysmorphism and Kenny-Caffey syndrome. Although a missense mutation in Tbce has been associated with progressive distal motor neuronopathy in the pmn/pmn mice, no similar degenerative phenotype has been recognized in humans. We report on the identification of an early-onset and progressive neurodegenerative encephalopathy with distal spinal muscular atrophy resembling the phenotype of pmn/pmn mice and caused by biallelic TBCE mutations, with the c.464T>A (p.Ile155Asn) change occurring at the heterozygous/homozygous state in six affected subjects from four unrelated families originated from the same geographical area in Southern Italy. Western blot analysis of patient fibroblasts documented a reduced amount of TBCE, suggestive of rapid degradation of the mutant protein, similarly to what was observed in pmn/pmn fibroblasts. The impact of TBCE mutations on microtubule polymerization was determined using biochemical fractionation and analyzing the nucleation and growth of microtubules at the centrosome and extracentrosomal sites after treatment with nocodazole. Primary fibroblasts obtained from affected subjects displayed a reduced level of polymerized α-tubulin, similarly to tail fibroblasts of pmn/pmn mice. Moreover, markedly delayed microtubule re-polymerization and abnormal mitotic spindles with disorganized microtubule arrangement were also documented. Although loss of function of TBCE has been documented to impact multiple developmental processes, the present findings provide evidence that hypomorphic TBCE mutations primarily drive neurodegeneration.


Subject(s)
Brain Diseases/complications , Brain Diseases/genetics , Molecular Chaperones/genetics , Muscular Atrophy, Spinal/complications , Muscular Atrophy, Spinal/genetics , Mutation/genetics , Adolescent , Age of Onset , Animals , Child , Female , Fibroblasts , Heterozygote , Homozygote , Humans , Infant , Infant, Newborn , Italy , Male , Mice , Microtubules/drug effects , Microtubules/metabolism , Microtubules/pathology , Molecular Chaperones/metabolism , Nocodazole/pharmacology , Spindle Apparatus/metabolism , Spindle Apparatus/pathology , Tubulin/metabolism , Young Adult
10.
Mol Neurodegener ; 11(1): 43, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27277231

ABSTRACT

BACKGROUND: Pathological Golgi fragmentation represents a constant pre-clinical feature of many neurodegenerative diseases including amyotrophic lateral sclerosis (ALS) but its molecular mechanisms remain hitherto unclear. RESULTS: Here, we show that the severe Golgi fragmentation in transgenic mutant SOD1(G85R) and SOD1(G93A) mouse motor neurons is associated with defective polymerization of Golgi-derived microtubules, loss of the COPI coat subunit ß-COP, cytoplasmic dispersion of the Golgi tether GM130, strong accumulation of the ER-Golgi v-SNAREs GS15 and GS28 as well as tubular/vesicular Golgi fragmentation. Data mining, transcriptomic and protein analyses demonstrate that both SOD1 mutants cause early presymptomatic and rapidly progressive up-regulation of the microtubule-destabilizing proteins Stathmins 1 and 2. Remarkably, mutant SOD1-triggered Golgi fragmentation and Golgi SNARE accumulation are recapitulated by Stathmin 1/2 overexpression but completely rescued by Stathmin 1/2 knockdown or the microtubule-stabilizing drug Taxol. CONCLUSIONS: We conclude that Stathmin-triggered microtubule destabilization mediates Golgi fragmentation in mutant SOD1-linked ALS and potentially also in related motor neuron diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Golgi Apparatus/pathology , Microtubules/pathology , Motor Neurons/pathology , Stathmin/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Immunoblotting , Immunohistochemistry , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron, Transmission , Motor Neurons/metabolism , Superoxide Dismutase-1/genetics
11.
Front Neurosci ; 9: 448, 2015.
Article in English | MEDLINE | ID: mdl-26696811

ABSTRACT

Pathological alterations of the Golgi apparatus, such as its fragmentation represent an early pre-clinical feature of many neurodegenerative diseases and have been widely studied in the motor neuron disease amyotrophic lateral sclerosis (ALS). Yet, the underlying molecular mechanisms have remained cryptic. In principle, Golgi fragmentation may result from defects in three major classes of proteins: structural Golgi proteins, cytoskeletal proteins and molecular motors, as well as proteins mediating transport to and through the Golgi. Here, we present the different mechanisms that may underlie Golgi fragmentation in animal and cellular models of ALS linked to mutations in SOD1, TARDBP (TDP-43), VAPB, and C9Orf72 and we propose a novel one based on findings in progressive motor neuronopathy (pmn) mice. These mice are mutated in the TBCE gene encoding the cis-Golgi localized tubulin-binding cofactor E, one of five chaperones that assist in tubulin folding and microtubule polymerization. Loss of TBCE leads to alterations in Golgi microtubules, which in turn impedes on the maintenance of the Golgi architecture. This is due to down-regulation of COPI coat components, dispersion of Golgi tethers and strong accumulation of ER-Golgi SNAREs. These effects are partially rescued by the GTPase ARF1 through recruitment of TBCE to the Golgi. We hypothesize that defects in COPI vesicles, microtubules and their interaction may also underlie Golgi fragmentation in human ALS linked to other mutations, spinal muscular atrophy (SMA), and related motor neuron diseases. We also discuss the functional relevance of pathological Golgi alterations, in particular their potential causative, contributory, or compensatory role in the degeneration of motor neuron cell bodies, axons and synapses.

12.
Neurobiol Dis ; 82: 269-280, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26107889

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a severe and incurable neurodegenerative disease. Human motor neurons generated from induced pluripotent stem cells (iPSc) offer new perspectives for disease modeling and drug testing in ALS. In standard iPSc-derived cultures, however, the two major phenotypic alterations of ALS--degeneration of motor neuron cell bodies and axons--are often obscured by cell body clustering, extensive axon criss-crossing and presence of unwanted cell types. Here, we succeeded in isolating 100% pure and standardized human motor neurons by a novel FACS double selection based on a p75(NTR) surface epitope and an HB9::RFP lentivirus reporter. The p75(NTR)/HB9::RFP motor neurons survive and grow well without forming clusters or entangled axons, are electrically excitable, contain ALS-relevant motor neuron subtypes and form functional connections with co-cultured myotubes. Importantly, they undergo rapid and massive cell death and axon degeneration in response to mutant SOD1 astrocytes. These data demonstrate the potential of FACS-isolated human iPSc-derived motor neurons for improved disease modeling and drug testing in ALS and related motor neuron diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , Flow Cytometry/methods , Induced Pluripotent Stem Cells , Motor Neurons , Adult , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Astrocytes/pathology , Astrocytes/physiology , Axons/pathology , Axons/physiology , Cell Survival , Cells, Cultured , Child , Coculture Techniques , Genes, Reporter , Humans , Induced Pluripotent Stem Cells/physiology , Lentivirus , Motor Neurons/pathology , Motor Neurons/physiology , Mutation , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
14.
Hum Mol Genet ; 23(22): 5961-75, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-24951541

ABSTRACT

Golgi fragmentation is an early hallmark of many neurodegenerative diseases but its pathophysiological relevance and molecular mechanisms are unclear. We here demonstrate severe and progressive Golgi fragmentation in motor neurons of progressive motor neuronopathy (pmn) mice due to loss of the Golgi-localized tubulin-binding cofactor E (TBCE). Loss of TBCE in mutant pmn and TBCE-depleted motor neuron cultures causes defects in Golgi-derived microtubules, as expected, but surprisingly also reduced levels of COPI subunits, decreased recruitment of tethering factors p115/GM130 and impaired Golgi SNARE-mediated vesicle fusion. Conversely, ARF1, which stimulates COPI vesicle formation, enhances the recruitment of TBCE to the Golgi, increases polymerization of Golgi-derived microtubules and rescues TBCE-linked Golgi fragmentation. These data indicate an ARF1/TBCE-mediated cross-talk that coordinates COPI formation and tubulin polymerization at the Golgi. We conclude that interruption of this cross-talk causes Golgi fragmentation in pmn mice and hypothesize that similar mechanisms operate in human amyotrophic lateral sclerosis and spinal muscular atrophy.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Amyotrophic Lateral Sclerosis/metabolism , COP-Coated Vesicles/metabolism , Golgi Apparatus/metabolism , Molecular Chaperones/metabolism , Muscular Atrophy, Spinal/metabolism , Tubulin/metabolism , ADP-Ribosylation Factor 1/genetics , Amyotrophic Lateral Sclerosis/genetics , Animals , COP-Coated Vesicles/genetics , Coat Protein Complex I/metabolism , Disease Models, Animal , Golgi Apparatus/chemistry , Humans , Mice , Mice, Inbred C57BL , Molecular Chaperones/genetics , Motor Neurons/chemistry , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Polymerization , Signal Transduction , Tubulin/chemistry
15.
Proc Natl Acad Sci U S A ; 110(1): 348-53, 2013 Jan 02.
Article in English | MEDLINE | ID: mdl-23248270

ABSTRACT

In healthy adults, activation of γ-aminobutyric acid (GABA)(A) and glycine receptors inhibits neurons as a result of low intracellular chloride concentration ([Cl(-)](i)), which is maintained by the potassium-chloride cotransporter KCC2. A reduction of KCC2 expression or function is implicated in the pathogenesis of several neurological disorders, including spasticity and chronic pain following spinal cord injury (SCI). Given the critical role of KCC2 in regulating the strength and robustness of inhibition, identifying tools that may increase KCC2 function and, hence, restore endogenous inhibition in pathological conditions is of particular importance. We show that activation of 5-hydroxytryptamine (5-HT) type 2A receptors to serotonin hyperpolarizes the reversal potential of inhibitory postsynaptic potentials (IPSPs), E(IPSP), in spinal motoneurons, increases the cell membrane expression of KCC2 and both restores endogenous inhibition and reduces spasticity after SCI in rats. Up-regulation of KCC2 function by targeting 5-HT(2A) receptors, therefore, has therapeutic potential in the treatment of neurological disorders involving altered chloride homeostasis. However, these receptors have been implicated in several psychiatric disorders, and their effects on pain processing are controversial, highlighting the need to further investigate the potential systemic effects of specific 5-HT(2A)R agonists, such as (4-bromo-3,6-dimethoxybenzocyclobuten-1-yl)methylamine hydrobromide (TCB-2).


Subject(s)
Gene Expression Regulation/drug effects , Inhibitory Postsynaptic Potentials/physiology , Motor Neurons/metabolism , Muscle Spasticity/drug therapy , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin/pharmacology , Spinal Cord Injuries/complications , Symporters/metabolism , Animals , Blotting, Western , Bridged Bicyclo Compounds/pharmacology , Chlorides/metabolism , H-Reflex , Immunohistochemistry , Methylamines/pharmacology , Muscle Spasticity/etiology , Rats , Serotonin/metabolism , Serotonin 5-HT2 Receptor Agonists/pharmacology , K Cl- Cotransporters
16.
Mol Cell Neurosci ; 46(2): 409-18, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21111824

ABSTRACT

Exosomes are microvesicles released into the extracellular medium upon fusion to the plasma membrane of endosomal intermediates called multivesicular bodies. They represent ways for discarding proteins and metabolites and also for intercellular transfer of proteins and RNAs. In the nervous system, it has been hypothesized that exosomes might be involved in the normal physiology of the synapse and possibly allow the trans-synaptic propagation of pathogenic proteins throughout the tissue. As a first step to validate this concept, we used biochemical and morphological approaches to demonstrate that mature cortical neurons in culture do indeed secrete exosomes. Using electron microscopy, we observed exosomes being released from somato-dendritic compartments. The endosomal origin of exosomes was demonstrated by showing that the C-terminal domain of tetanus toxin specifically endocytosed by neurons and accumulating inside multivesicular bodies, is released in the extracellular medium in association with exosomes. Finally, we found that exosomal release is modulated by glutamatergic synaptic activity, suggesting that this process might be part of normal synaptic physiology. Thus, our study paves the way towards the demonstration that exosomes take part in the physiology of the normal and pathological nervous system.


Subject(s)
Exosomes/metabolism , Neurons/metabolism , Synapses/metabolism , Animals , Blotting, Western , Cell Differentiation , Cells, Cultured , Exosomes/ultrastructure , Glutamine/metabolism , Microscopy, Electron, Transmission , Neurons/ultrastructure , Rats , Synapses/ultrastructure
17.
Curr Opin Neurobiol ; 18(3): 284-91, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18725296

ABSTRACT

Cell death plays an important role both in shaping the developing nervous system and in neurological disease and traumatic injury. In spite of their name, death receptors can trigger either cell death or survival and growth. Recent studies implicate five death receptors--Fas/CD95, TNFR1 (tumor necrosis factor receptor-1), p75NTR (p75 neurotrophin receptor), DR4, and DR5 (death receptors-4 and -5)--in different aspects of neural development or degeneration. Their roles may be neuroprotective in models of Parkinson's disease, or pro-apoptotic in ALS and stroke. Such different outcomes probably reflect the diversity of transcriptional and posttranslational signaling pathways downstream of death receptors in neurons and glia.


Subject(s)
Nervous System Physiological Phenomena , Receptors, Death Domain/metabolism , Signal Transduction/physiology , Animals
18.
Stem Cells ; 26(10): 2564-75, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18635866

ABSTRACT

Stem cell-based therapies hold therapeutic promise for degenerative motor neuron diseases, such as amyotrophic lateral sclerosis, and for spinal cord injury. Fetal neural progenitors present less risk of tumor formation than embryonic stem cells but inefficiently differentiate into motor neurons, in line with their low expression of motor neuron-specific transcription factors and poor response to soluble external factors. To overcome this limitation, we genetically engineered fetal rat spinal cord neurospheres to express the transcription factors HB9, Nkx6.1, and Neurogenin2. Enforced expression of the three factors rendered neural precursors responsive to Sonic hedgehog and retinoic acid and directed their differentiation into cholinergic motor neurons that projected axons and formed contacts with cocultured myotubes. When transplanted in the injured adult rat spinal cord, a model of acute motor neuron degeneration, the engineered precursors transiently proliferated, colonized the ventral horn, expressed motor neuron-specific differentiation markers, and projected cholinergic axons in the ventral root. We conclude that genetic engineering can drive the differentiation of fetal neural precursors into motor neurons that efficiently engraft in the spinal cord. The strategy thus holds promise for cell replacement in motor neuron and related diseases. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Directed Molecular Evolution , Genetic Engineering , Motor Neurons/cytology , Motor Neurons/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Axons/metabolism , Biomarkers/metabolism , Cell Communication , Cell Differentiation , Cell Movement , Choline/metabolism , Coculture Techniques , Humans , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Organ Specificity , Rats , Spinal Cord Injuries/pathology , Spinal Nerve Roots/pathology , Stem Cell Transplantation , Transcription Factors/metabolism
19.
J Neurosci ; 27(33): 8779-89, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17699660

ABSTRACT

Axonal degeneration represents one of the earliest pathological features in motor neuron diseases. We here studied the underlying molecular mechanisms in progressive motor neuronopathy (pmn) mice mutated in the tubulin-specific chaperone TBCE. We demonstrate that TBCE is a peripheral membrane-associated protein that accumulates at the Golgi apparatus. In pmn mice, TBCE is destabilized and disappears from the Golgi apparatus of motor neurons, and microtubules are lost in distal axons. The axonal microtubule loss proceeds retrogradely in parallel with the axonal dying back process. These degenerative changes are inhibited in a dose-dependent manner by transgenic TBCE complementation that restores TBCE expression at the Golgi apparatus. In cultured motor neurons, the pmn mutation, interference RNA-mediated TBCE depletion, and brefeldin A-mediated Golgi disruption all compromise axonal tubulin routing. We conclude that motor axons critically depend on axonal tubulin routing from the Golgi apparatus, a process that involves TBCE and possibly other tubulin chaperones.


Subject(s)
Golgi Apparatus/metabolism , Molecular Chaperones/physiology , Motor Neuron Disease/pathology , Motor Neurons/ultrastructure , Nerve Degeneration/pathology , Tubulin/metabolism , Animals , Cells, Cultured , Disease Progression , Embryo, Mammalian , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Golgi Apparatus/drug effects , Green Fluorescent Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Microtubules/metabolism , Microtubules/pathology , Microtubules/ultrastructure , Molecular Chaperones/genetics , Motor Neurons/drug effects , Motor Neurons/pathology , RNA, Messenger/biosynthesis , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction/methods , Spinal Cord/metabolism , Spinal Cord/pathology
20.
Am J Hum Genet ; 81(1): 1-16, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17564959

ABSTRACT

Charcot-Marie-Tooth (CMT) disorders are a clinically and genetically heterogeneous group of hereditary motor and sensory neuropathies characterized by muscle weakness and wasting, foot and hand deformities, and electrophysiological changes. The CMT4H subtype is an autosomal recessive demyelinating form of CMT that was recently mapped to a 15.8-Mb region at chromosome 12p11.21-q13.11, in two consanguineous families of Mediterranean origin, by homozygosity mapping. We report here the identification of mutations in FGD4, encoding FGD4 or FRABIN (FGD1-related F-actin binding protein), in both families. FRABIN is a GDP/GTP nucleotide exchange factor (GEF), specific to Cdc42, a member of the Rho family of small guanosine triphosphate (GTP)-binding proteins (Rho GTPases). Rho GTPases play a key role in regulating signal-transduction pathways in eukaryotes. In particular, they have a pivotal role in mediating actin cytoskeleton changes during cell migration, morphogenesis, polarization, and division. Consistent with these reported functions, expression of truncated FRABIN mutants in rat primary motoneurons and rat Schwann cells induced significantly fewer microspikes than expression of wild-type FRABIN. To our knowledge, this is the first report of mutations in a Rho GEF protein being involved in CMT.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Genes, Recessive , Guanine Nucleotide Exchange Factors/genetics , Microfilament Proteins/genetics , Amino Acid Sequence , Animals , Chromosomes, Human, Pair 12/genetics , Gene Expression , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Humans , Microfilament Proteins/analysis , Molecular Sequence Data , Mutation , Pedigree , Physical Chromosome Mapping , Rats , Schwann Cells/enzymology , rho GTP-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...