Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Hum Mol Genet ; 28(4): 584-597, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30335140

ABSTRACT

Mutations in the Tre2/Bub2/Cdc16 (TBC)1 domain family member 24 (TBC1D24) gene are associated with a range of inherited neurological disorders, from drug-refractory lethal epileptic encephalopathy and DOORS syndrome (deafness, onychodystrophy, osteodystrophy, mental retardation, seizures) to non-syndromic hearing loss. TBC1D24 has been implicated in neuronal transmission and maturation, although the molecular function of the gene and the cause of the apparently complex disease spectrum remain unclear. Importantly, heterozygous TBC1D24 mutation carriers have also been reported with seizures, suggesting that haploinsufficiency for TBC1D24 is significant clinically. Here we have systematically investigated an allelic series of disease-associated mutations in neurons alongside a new mouse model to investigate the consequences of TBC1D24 haploinsufficiency to mammalian neurodevelopment and synaptic physiology. The cellular studies reveal that disease-causing mutations that disrupt either of the conserved protein domains in TBC1D24 are implicated in neuronal development and survival and are likely acting as loss-of-function alleles. We then further investigated TBC1D24 haploinsufficiency in vivo and demonstrate that TBC1D24 is also crucial for normal presynaptic function: genetic disruption of Tbc1d24 expression in the mouse leads to an impairment of endocytosis and an enlarged endosomal compartment in neurons with a decrease in spontaneous neurotransmission. These data reveal the essential role for TBC1D24 at the mammalian synapse and help to define common synaptic mechanisms that could underlie the varied effects of TBC1D24 mutations in neurological disease.


Subject(s)
Carrier Proteins/genetics , Craniofacial Abnormalities/genetics , Epilepsy/genetics , Hand Deformities, Congenital/genetics , Hearing Loss, Sensorineural/genetics , Intellectual Disability/genetics , Nails, Malformed/genetics , Seizures/genetics , Amino Acid Sequence/genetics , Animals , Craniofacial Abnormalities/physiopathology , Disease Models, Animal , Endocytosis/genetics , Epilepsy/physiopathology , Exome/genetics , GTPase-Activating Proteins , Gene Expression Regulation , Hand Deformities, Congenital/physiopathology , Haploinsufficiency , Hearing Loss, Sensorineural/physiopathology , Humans , Intellectual Disability/physiopathology , Membrane Proteins , Mice , Mutation , Nails, Malformed/physiopathology , Nerve Tissue Proteins , Neuronal Plasticity/genetics , Neurons/metabolism , Neurons/pathology , Pedigree , Seizures/physiopathology
2.
Brain ; 143(6): 1731-1745, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32437528

ABSTRACT

Autoantibodies against leucine-rich glioma inactivated 1 (LGI1) are found in patients with limbic encephalitis and focal seizures. Here, we generate patient-derived monoclonal antibodies (mAbs) against LGI1. We explore their sequences and binding characteristics, plus their pathogenic potential using transfected HEK293T cells, rodent neuronal preparations, and behavioural and electrophysiological assessments in vivo after mAb injections into the rodent hippocampus. In live cell-based assays, LGI1 epitope recognition was examined with patient sera (n = 31), CSFs (n = 11), longitudinal serum samples (n = 15), and using mAbs (n = 14) generated from peripheral B cells of two patients. All sera and 9/11 CSFs bound both the leucine-rich repeat (LRR) and the epitempin repeat (EPTP) domains of LGI1, with stable ratios of LRR:EPTP antibody levels over time. By contrast, the mAbs derived from both patients recognized either the LRR or EPTP domain. mAbs against both domain specificities showed varied binding strengths, and marked genetic heterogeneity, with high mutation frequencies. LRR-specific mAbs recognized LGI1 docked to its interaction partners, ADAM22 and ADAM23, bound to rodent brain sections, and induced internalization of the LGI1-ADAM22/23 complex in both HEK293T cells and live hippocampal neurons. By contrast, few EPTP-specific mAbs bound to rodent brain sections or ADAM22/23-docked LGI1, but all inhibited the docking of LGI1 to ADAM22/23. After intrahippocampal injection, and by contrast to the LRR-directed mAbs, the EPTP-directed mAbs showed far less avid binding to brain tissue and were consistently detected in the serum. Post-injection, both domain-specific mAbs abrogated long-term potentiation induction, and LRR-directed antibodies with higher binding strengths induced memory impairment. Taken together, two largely dichotomous populations of LGI1 mAbs with distinct domain binding characteristics exist in the affinity matured peripheral autoantigen-specific memory pools of individuals, both of which have pathogenic potential. In human autoantibody-mediated diseases, the detailed characterization of patient mAbs provides a valuable method to dissect the molecular mechanisms within polyclonal populations.


Subject(s)
Antibodies, Monoclonal/metabolism , Autoantibodies/immunology , Intracellular Signaling Peptides and Proteins/immunology , ADAM Proteins/metabolism , Animals , Antibodies, Monoclonal/immunology , Autoantibodies/blood , Autoantibodies/metabolism , Autoantigens/metabolism , Brain/metabolism , Epitopes/immunology , HEK293 Cells , Hippocampus/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Limbic Encephalitis/blood , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Protein Binding/immunology , Protein Domains/immunology
3.
Cereb Cortex ; 29(5): 2148-2159, 2019 05 01.
Article in English | MEDLINE | ID: mdl-29850799

ABSTRACT

Synaptosomal associated protein 25 kDa (SNAP25) is an essential component of the SNARE complex regulating synaptic vesicle fusion. SNAP25 deficiency has been implicated in a variety of cognitive disorders. We ablated SNAP25 from selected neuronal populations by generating a transgenic mouse (B6-Snap25tm3mcw (Snap25-flox)) with LoxP sites flanking exon5a/5b. In the presence of Cre-recombinase, Snap25-flox is recombined to a truncated transcript. Evoked synaptic vesicle release is severely reduced in Snap25 conditional knockout (cKO) neurons as shown by live cell imaging of synaptic vesicle fusion and whole cell patch clamp recordings in cultured hippocampal neurons. We studied Snap25 cKO in subsets of cortical projection neurons in vivo (L5-Rbp4-Cre; L6-Ntsr1-Cre; L6b-Drd1a-Cre). cKO neurons develop normal axonal projections, but axons are not maintained appropriately, showing signs of swelling, fragmentation and eventually complete absence. Onset and progression of degeneration are dependent on the neuron type, with L5 cells showing the earliest and most severe axonal loss. Ultrastructural examination revealed that cKO neurites contain autophagosome/lysosome-like structures. Markers of inflammation such as Iba1 and lipofuscin are increased only in adult cKO cortex. Snap25 cKO can provide a model to study genetic interactions with environmental influences in several disorders.


Subject(s)
Brain/growth & development , Brain/pathology , Neurons/pathology , Neurons/physiology , Synaptosomal-Associated Protein 25/physiology , Animals , Axons/pathology , Axons/physiology , Axons/ultrastructure , Brain/ultrastructure , Female , Male , Mice, Knockout , Neurons/ultrastructure , Synaptic Transmission , Synaptic Vesicles
4.
Neuronal Signal ; 7(1): NS20220086, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36687366

ABSTRACT

The amyloid hypothesis, which proposes that accumulation of the peptide amyloid-ß at synapses is the key driver of Alzheimer's disease (AD) pathogenesis, has been the dominant idea in the field of Alzheimer's research for nearly 30 years. Recently, however, serious doubts about its validity have emerged, largely motivated by disappointing results from anti-amyloid therapeutics in clinical trials. As a result, much of the AD research effort has shifted to understanding the roles of a variety of other entities implicated in pathogenesis, such as microglia, astrocytes, apolipoprotein E and several others. All undoubtedly play an important role, but the nature of this has in many cases remained unclear, partly due to their pleiotropic functions. Here, we propose that all of these AD-related entities share at least one overlapping function, which is the local regulation of amyloid-ß levels, and that this may be critical to their role in AD pathogenesis. We also review what is currently known of the actions of amyloid-ß at the synapse in health and disease, and consider in particular how it might interact with the key AD-associated protein tau in the disease setting. There is much compelling evidence in support of the amyloid hypothesis; rather than detract from this, the implication of many disparate AD-associated cell types, molecules and processes in the regulation of amyloid-ß levels may lend further support.

5.
Front Cell Neurosci ; 15: 782768, 2021.
Article in English | MEDLINE | ID: mdl-34955753

ABSTRACT

Homeostatic synaptic plasticity (HSP) regulates synaptic strength both pre- and postsynaptically to ensure stability and efficient information transfer in neural networks. A number of neurological diseases have been associated with deficits in HSP, particularly diseases characterised by episodic network instability such as migraine and epilepsy. Recently, it has become apparent that HSP also plays a role in many neurodegenerative diseases. In this mini review, we present an overview of the evidence linking HSP to each of the major neurodegenerative diseases, finding that HSP changes in each disease appear to belong to one of three broad functional categories: (1) deficits in HSP at degenerating synapses that contribute to pathogenesis or progression; (2) HSP induced in a heterosynaptic or cell non-autonomous manner to support the function of networks of which the degenerating synapses or cells are part; and (3) induction of HSP within the degenerating population of synapses to preserve function and to resist the impact of synapse loss. Understanding the varied manifestations of HSP in neurodegeneration will not only aid understanding mechanisms of disease but could also inspire much-needed novel approaches to therapy.

6.
Cell Rep ; 36(9): 109638, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34469725

ABSTRACT

In Alzheimer's disease, soluble oligomers of the amyloid-ß peptide (Aßo) trigger a cascade of events that includes abnormal hyperphosphorylation of the protein tau, which is essential for pathogenesis. However, the mechanistic link between these two key pathological proteins remains unclear. Using hippocampal slices, we show here that an Aßo-mediated increase in glutamate release probability causes enhancement of synaptically evoked N-methyl-d-aspartate subtype glutamate receptor (NMDAR)-dependent long-term depression (LTD). We also find that elevated glutamate release probability is required for Aßo-induced pathological hyperphosphorylation of tau, which is likewise NMDAR dependent. Finally, we show that chronic, repeated chemical or optogenetic induction of NMDAR-dependent LTD alone is sufficient to cause tau hyperphosphorylation without Aßo. Together, these results support a possible causal chain in which Aßo increases glutamate release probability, thus leading to enhanced LTD induction, which in turn drives hyperphosphorylation of tau. Our data identify a mechanistic pathway linking the two critical pathogenic proteins of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/toxicity , Hippocampus/drug effects , Long-Term Synaptic Depression , Peptide Fragments/toxicity , Synapses/drug effects , tau Proteins/metabolism , Alzheimer Disease/physiopathology , Animals , Excitatory Postsynaptic Potentials/drug effects , Female , Glutamic Acid/metabolism , Hippocampus/metabolism , Hippocampus/physiopathology , In Vitro Techniques , Male , Mice, Inbred C57BL , Phosphorylation , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism
7.
Front Cell Neurosci ; 14: 564081, 2020.
Article in English | MEDLINE | ID: mdl-33240043

ABSTRACT

Non-evoked miniature release of neurotransmitters is increasingly recognized as playing an important role in neural function and is implicated in synaptic plasticity, metaplasticity, and homeostasis. Spontaneous miniature release events (minis) are usually measured electrophysiologically by recording the miniature postsynaptic currents (mEPSCs) that they evoke. However, this indirect technique can be confounded by changes within the postsynaptic neuron. Here, using the fluorescent probe SynaptopHluorin 2×, we have developed an optical method for the measurement of minis that enables direct assessment of release events. We use the technique to reveal that the frequency of minis following incubation of hippocampal neurons with Amyloid ß oligomers (Aßo) is increased. Electrophysiological mEPSC recordings obtained under the same conditions report a decrease in frequency, with the discrepancy likely due to Aßo-induced changes in quantal size. Optical quantal analysis of minis may therefore have a role in the study of minis in both normal physiology and disease, as it can circumvent potential confounds caused by postsynaptic changes.

9.
Pract Neurol ; 8(5): 303-10, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18796585

ABSTRACT

In this article we summarise nervous system histology in health and disease and acquaint the reader with developments in the staining techniques that are in current use, particularly immunostains. Although clinicians do not need to know the details of stain appearances, some familiarity with these aspects of neuropathology is invaluable in interpreting the reports they receive from the laboratory, as well as reminding them of the amazing beauty of the central nervous system's microscopic structure.


Subject(s)
Brain/pathology , Histology/trends , Neurons/pathology , Pathology/methods , Staining and Labeling/methods , Biomarkers/analysis , Biomarkers/metabolism , Brain/metabolism , Brain/physiopathology , Coloring Agents/standards , Humans , Immunohistochemistry/methods , Immunohistochemistry/trends , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Pathology/standards , Staining and Labeling/trends
11.
Sci Signal ; 11(558)2018 11 27.
Article in English | MEDLINE | ID: mdl-30482851

ABSTRACT

Acidic organelles, such as endosomes and lysosomes, store Ca2+ that is released in response to intracellular increases in the second messenger nicotinic acid adenine dinucleotide phosphate (NAADP). In neurons, NAADP and Ca2+ signaling contribute to synaptic plasticity, a process of activity-dependent long-term potentiation (LTP) [or, alternatively, long-term depression (LTD)] of synaptic strength and neuronal transmission that is critical for neuronal function and memory formation. We explored the function of and mechanisms regulating acidic Ca2+ store signaling in murine hippocampal neurons. We found that metabotropic glutamate receptor 1 (mGluR1) was coupled to NAADP signaling that elicited Ca2+ release from acidic stores. In turn, this released Ca2+-mediated mGluR1-dependent LTP by transiently inhibiting SK-type K+ channels, possibly through the activation of protein phosphatase 2A. Genetically removing two-pore channels (TPCs), which are endolysosomal-specific ion channels, switched the polarity of plasticity from LTP to LTD, indicating the importance of specific receptor store coupling and providing mechanistic insight into how mGluR1 can produce both synaptic potentiation and synaptic depression.


Subject(s)
Calcium Channels/physiology , Calcium Signaling , Calcium/metabolism , Hippocampus/physiology , Long-Term Potentiation , NADP/analogs & derivatives , Receptors, Metabotropic Glutamate/metabolism , Animals , Cells, Cultured , Hippocampus/drug effects , Male , Mice , Mice, Knockout , NADP/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats , Rats, Wistar
12.
Cell Rep ; 21(2): 341-350, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-29020622

ABSTRACT

Voltage-dependent Ca2+ channels (VGCC) represent the principal source of Ca2+ ions driving evoked neurotransmitter release at presynaptic boutons. In mammals, presynaptic Ca2+ influx is mediated mainly via P/Q-type and N-type VGCC, which differ in their properties. Changes in their relative contributions tune neurotransmission both during development and in Hebbian plasticity. However, whether this represents a functional motif also present in other forms of activity-dependent regulation is unknown. Here, we study the role of VGCC in homeostatic plasticity (HSP) in mammalian hippocampal neurons using optical techniques. We find that changes in evoked Ca2+ currents specifically through P/Q-type, but not N-type, VGCC mediate bidirectional homeostatic regulation of both neurotransmitter release efficacy and the size of the major synaptic vesicle pools. Selective dependence of HSP on P/Q-type VGCC in mammalian terminals has important implications for phenotypes associated with P/Q-type channelopathies, including migraine and epilepsy.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Channels, P-Type/metabolism , Homeostasis , Neuronal Plasticity , Neurons/metabolism , Presynaptic Terminals/metabolism , Animals , Cells, Cultured , Hippocampus/cytology , Mice , Neurons/physiology , Presynaptic Terminals/physiology , Rats , Synaptic Vesicles/metabolism
13.
J Neurosci ; 23(5): 1631-7, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12629167

ABSTRACT

The robotic mouse is an autosomal dominant mutant that arose from a large-scale chemical mutagenesis program. It has a jerky, ataxic gait and develops adult-onset Purkinje cell loss in the cerebellum in a striking region-specific pattern, as well as cataracts. Genetic and physical mapping of the disease locus led to the identification of a missense mutation in a highly conserved region of Af4, a putative transcription factor that has been previously implicated in leukemogenesis. We demonstrate that Af4 is specifically expressed in Purkinje cells, and we hypothesize that the expression of mutant Af4 leads to neurodegeneration. This function was not identified through knock-out studies, highlighting the power of phenotype-driven mutagenesis in the mouse to identify new pathways involved in neurological disease.


Subject(s)
Cataract/genetics , Cerebellar Ataxia/genetics , Cerebellum/pathology , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Purkinje Cells/pathology , Amino Acid Sequence , Animals , Antigens, CD/biosynthesis , Cell Count , Cerebellar Ataxia/pathology , Conserved Sequence , DNA-Binding Proteins/biosynthesis , Disease Models, Animal , Disease Progression , Flow Cytometry , Genes, Dominant , Mice , Mice, Neurologic Mutants , Molecular Sequence Data , Mutagenesis , Nuclear Proteins/biosynthesis , Organ Specificity/genetics , Physical Chromosome Mapping , Point Mutation , Purkinje Cells/metabolism , Sequence Homology, Amino Acid , Thymus Gland/metabolism , Thymus Gland/pathology
14.
Neurol Res Int ; 2015: 345285, 2015.
Article in English | MEDLINE | ID: mdl-25653875

ABSTRACT

Parkinson's disease (PD) is the world's second most common neurodegenerative disease and most common movement disorder. Characterised by a loss of dopaminergic neurons and the development of intraneuronal inclusions known as Lewy bodies, it has classically been thought of as a cell-autonomous disease. However, in 2008, two groups reported the startling observation of Lewy bodies within embryonic neuronal grafts transplanted into PD patients little more than a decade previously, suggesting that PD pathology can be propagated to neighbouring cells and calling basic assumptions of our understanding of the disease into question. Subsequent research has largely served to confirm this interpretation, pointing towards a prion-like intercellular transfer of misfolded α-synuclein, the main component of Lewy bodies, as central to PD. This shift in thinking offers a revolutionary approach to PD treatment, potentially enabling a transition from purely symptomatic therapy to direct targeting of the pathology that drives disease progression. In this short review, we appraise current experimental support for PD as a prion-like disease, whilst highlighting areas of controversy or inconsistency which must be resolved. We also offer a brief discussion of the therapeutic implications of these discoveries.

16.
Neuropharmacology ; 61(8): 1345-53, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21856316

ABSTRACT

The effects of the major schizophrenia susceptibility gene disease DTNBP1 on disease risk are likely to be mediated through changes in expression level of the gene product, dysbindin-1. How such changes might influence pathogenesis is, however, unclear. One possible mechanism is suggested by recent work establishing a link between altered dysbindin-1 expression and changes in surface levels of N-methyl-d-aspartate receptors (NMDAR), although neither the precise nature of this relationship, nor the mechanism underlying it, are understood. Using organotypic slices of rat hippocampus, we show that increased expression of dysbindin-1A in pyramidal neurons causes a severe and selective hypofunction of NMDARs and blocks induction of LTP. Cell surface, but not cytoplasmic, expression of the NR1 subunit of the NMDAR is decreased, suggesting dysregulation of NMDAR trafficking and, consistent with this, pharmacological inhibition of clathrin-dependent endocytosis is sufficient to reverse the deficit in NMDAR signaling. These results support the idea that the level of the NMDAR at the plasma membrane is modulated by changes in dysbindin-1 expression and offer further insight into the role of dysbindin-1 at an important cellular pathway implicated in schizophrenia.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation/physiology , Hippocampus/cytology , Neurons/metabolism , Signal Transduction/physiology , Animals , Bacterial Proteins/genetics , Biophysics , Cells, Cultured , Clathrin/pharmacology , Dysbindin , Dystrophin-Associated Proteins , Electric Stimulation , Embryo, Mammalian , Endocytosis/drug effects , Endocytosis/physiology , Excitatory Amino Acid Agonists/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Luminescent Proteins/genetics , Male , Mice , N-Methylaspartate/pharmacology , Neurons/drug effects , Organ Culture Techniques , Patch-Clamp Techniques , Protein Transport/drug effects , Protein Transport/genetics , Rats , Rats, Wistar , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Transfection/methods , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
17.
Nat Rev Clin Oncol ; 6(5): 295-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19390556

ABSTRACT

BACKGROUND: A 37-year-old woman presented with a supratentorial cerebral mass, which was diagnosed histologically as a primitive neuroectodermal tumor. She had been treated for rectal adenocarcinoma 7 years previously. A family history revealed a young-onset colorectal carcinoma in the patient's father. INVESTIGATIONS: Immunohistochemical analysis for DNA mismatch repair proteins, germline mutation analysis of MSH2. DIAGNOSIS: Lynch syndrome with a heterozygous germline mutation in MSH2. MANAGEMENT: Debulking of the cerebral tumor, craniospinal axis radiotherapy, and genetic counseling of family.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Heterozygote , MutS Homolog 2 Protein/genetics , Mutation , Neuroectodermal Tumors, Primitive/genetics , Adult , DNA Mismatch Repair/genetics , DNA Mutational Analysis , Fatal Outcome , Female , Germ-Line Mutation , Humans , Immunohistochemistry , Neuroectodermal Tumors, Primitive/diagnosis , Neuroectodermal Tumors, Primitive/diagnostic imaging , Neuroectodermal Tumors, Primitive/radiotherapy , Pedigree , Radiography , Sequence Analysis, DNA
18.
Proc Natl Acad Sci U S A ; 104(7): 2431-6, 2007 Feb 13.
Article in English | MEDLINE | ID: mdl-17283335

ABSTRACT

The neuronal soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex is essential for synaptic vesicle exocytosis, but its study has been limited by the neonatal lethality of murine SNARE knockouts. Here, we describe a viable mouse line carrying a mutation in the b-isoform of neuronal SNARE synaptosomal-associated protein of 25 kDa (SNAP-25). The causative I67T missense mutation results in increased binding affinities within the SNARE complex, impaired exocytotic vesicle recycling and granule exocytosis in pancreatic beta-cells, and a reduction in the amplitude of evoked cortical excitatory postsynaptic potentials. The mice also display ataxia and impaired sensorimotor gating, a phenotype which has been associated with psychiatric disorders in humans. These studies therefore provide insights into the role of the SNARE complex in both diabetes and psychiatric disease.


Subject(s)
Ataxia/genetics , Exocytosis/genetics , Gait Disorders, Neurologic/genetics , Mutation, Missense , Synaptic Vesicles/genetics , Synaptosomal-Associated Protein 25/genetics , Alcoholic Intoxication , Animals , Diabetes Mellitus/etiology , Genes, Dominant , Insulin-Secreting Cells , Mental Disorders/etiology , Mice , Mice, Mutant Strains , Models, Animal , SNARE Proteins/physiology , Synaptosomal-Associated Protein 25/physiology
19.
Mol Cell Neurosci ; 21(1): 114-25, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12359155

ABSTRACT

We have identified and characterized a new peripheral myelin protein 22 (Pmp22) mouse mutant. The mutation results in a serine to threonine amino acid substitution at residue 72, which is a hot spot for mutation in human PMP22, leading to the peripheral neuropathy Dejerine-Sottas syndrome. We have previously described two other Pmp22 mutants, providing an allelic series for gene function analysis. Pmp22 mutations generally lead to abnormal intracellular trafficking of Pmp22, and we show that each mutant protein in the allelic series has a unique pattern of intracellular localization in transfected cell lines. The mutant protein from the less severely affected mutants occurs in large aggregates, while the mutant protein from the most severely affected mutant occurs in a diffuse perinuclear pattern that largely colocalizes with wild-type protein. This suggests that large Pmp22 aggregates may be protective in this form of peripheral neuropathy.


Subject(s)
Mice, Neurologic Mutants/genetics , Myelin Proteins/genetics , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/metabolism , Alleles , Animals , COS Cells , Dimerization , Gene Expression/genetics , Genotype , HeLa Cells , Humans , Mice , Microscopy, Electron , Myelin Sheath/metabolism , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , Peripheral Nervous System Diseases/pathology , Phenotype , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Sciatic Nerve/ultrastructure , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL