Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Mol Ther ; 32(3): 800-817, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38243601

ABSTRACT

Hearing loss is a major health concern affecting millions of people worldwide with currently limited treatment options. In clarin-2-deficient Clrn2-/- mice, used here as a model of progressive hearing loss, we report synaptic auditory abnormalities in addition to the previously demonstrated defects of hair bundle structure and mechanoelectrical transduction. We sought an in-depth evaluation of viral-mediated gene delivery as a therapy for these hearing-impaired mice. Supplementation with either the murine Clrn2 or human CLRN2 genes preserved normal hearing in treated Clrn2-/- mice. Conversely, mutated forms of CLRN2, identified in patients with post-lingual moderate to severe hearing loss, failed to prevent hearing loss. The ectopic expression of clarin-2 successfully prevented the loss of stereocilia, maintained normal mechanoelectrical transduction, preserved inner hair cell synaptic function, and ensured near-normal hearing thresholds over time. Maximal hearing preservation was observed when Clrn2 was delivered prior to the loss of transducing stereocilia. Our findings demonstrate that gene therapy is effective for the treatment of post-lingual hearing impairment and age-related deafness associated with CLRN2 patient mutations.


Subject(s)
Hair Cells, Auditory , Hearing Loss , Humans , Animals , Mice , Hair Cells, Auditory/metabolism , Hearing , Hearing Loss/genetics , Hearing Loss/therapy , Stereocilia/metabolism , Dietary Supplements
2.
Front Neurol ; 13: 816534, 2022.
Article in English | MEDLINE | ID: mdl-35444606

ABSTRACT

The inner ear is responsible for both hearing and balance. These functions are dependent on the correct functioning of mechanosensitive hair cells, which convert sound- and motion-induced stimuli into electrical signals conveyed to the brain. During evolution of the inner ear, the major changes occurred in the hearing organ, whereas the structure of the vestibular organs remained constant in all vertebrates over the same period. Vestibular deficits are highly prevalent in humans, due to multiple intersecting causes: genetics, environmental factors, ototoxic drugs, infections and aging. Studies of deafness genes associated with balance deficits and their corresponding animal models have shed light on the development and function of these two sensory systems. Bilateral vestibular deficits often impair individual postural control, gaze stabilization, locomotion and spatial orientation. The resulting dizziness, vertigo, and/or falls (frequent in elderly populations) greatly affect patient quality of life. In the absence of treatment, prosthetic devices, such as vestibular implants, providing information about the direction, amplitude and velocity of body movements, are being developed and have given promising results in animal models and humans. Novel methods and techniques have led to major progress in gene therapies targeting the inner ear (gene supplementation and gene editing), 3D inner ear organoids and reprograming protocols for generating hair cell-like cells. These rapid advances in multiscale approaches covering basic research, clinical diagnostics and therapies are fostering interdisciplinary research to develop personalized treatments for vestibular disorders.

3.
Hum Genet ; 140(6): 915-931, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33496845

ABSTRACT

Deafness, the most frequent sensory deficit in humans, is extremely heterogeneous with hundreds of genes involved. Clinical and genetic analyses of an extended consanguineous family with pre-lingual, moderate-to-profound autosomal recessive sensorineural hearing loss, allowed us to identify CLRN2, encoding a tetraspan protein, as a new deafness gene. Homozygosity mapping followed by exome sequencing identified a 14.96 Mb locus on chromosome 4p15.32p15.1 containing a likely pathogenic missense variant in CLRN2 (c.494C > A, NM_001079827.2) segregating with the disease. Using in vitro RNA splicing analysis, we show that the CLRN2 c.494C > A variant leads to two events: (1) the substitution of a highly conserved threonine (uncharged amino acid) to lysine (charged amino acid) at position 165, p.(Thr165Lys), and (2) aberrant splicing, with the retention of intron 2 resulting in a stop codon after 26 additional amino acids, p.(Gly146Lysfs*26). Expression studies and phenotyping of newly produced zebrafish and mouse models deficient for clarin 2 further confirm that clarin 2, expressed in the inner ear hair cells, is essential for normal organization and maintenance of the auditory hair bundles, and for hearing function. Together, our findings identify CLRN2 as a new deafness gene, which will impact future diagnosis and treatment for deaf patients.


Subject(s)
Amino Acid Substitution , Chromosomes, Human, Pair 4/chemistry , Hair Cells, Auditory, Inner/metabolism , Hearing Loss, Sensorineural/genetics , Membrane Proteins/genetics , Point Mutation , Tetraspanins/genetics , Adult , Alleles , Animals , Base Sequence , Chromosome Mapping , Consanguinity , Female , Gene Expression , Genes, Recessive , Hair Cells, Auditory, Inner/pathology , Hearing Loss, Sensorineural/metabolism , Hearing Loss, Sensorineural/pathology , Humans , Male , Membrane Proteins/deficiency , Mice , Pedigree , Tetraspanins/deficiency , Exome Sequencing , Zebrafish
4.
J Biol Chem ; 294(37): 13755-13768, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31346033

ABSTRACT

Protection of neuronal homeostasis is a major goal in the management of neurodegenerative diseases. Microtubule-associated Ser/Thr kinase 2 (MAST2) inhibits neurite outgrowth, and its inhibition therefore represents a potential therapeutic strategy. We previously reported that a viral protein (G-protein from rabies virus) capable of interfering with protein-protein interactions between the PDZ domain of MAST2 and the C-terminal moieties of its cellular partners counteracts MAST2-mediated suppression of neurite outgrowth. Here, we designed peptides derived from the native viral protein to increase the affinity of these peptides for the MAST2-PDZ domain. Our strategy involved modifying the length and flexibility of the noninteracting sequence linking the two subsites anchoring the peptide to the PDZ domain. Three peptides, Neurovita1 (NV1), NV2, and NV3, were selected, and we found that they all had increased affinities for the MAST2-PDZ domain, with Kd values decreasing from 1300 to 60 nm, while target selectivity was maintained. A parallel biological assay evaluating neurite extension and branching in cell cultures revealed that the NV peptides gradually improved neural activity, with the efficacies of these peptides for stimulating neurite outgrowth mirroring their affinities for MAST2-PDZ. We also show that NVs can be delivered into the cytoplasm of neurons as a gene or peptide. In summary, our findings indicate that virus-derived peptides targeted to MAST2-PDZ stimulate neurite outgrowth in several neuron types, opening up promising avenues for potentially using NVs in the management of neurodegenerative diseases.


Subject(s)
Neurites/metabolism , Neuronal Outgrowth/drug effects , PDZ Domains/physiology , Central Nervous System Stimulants/metabolism , Humans , Induced Pluripotent Stem Cells , Microtubules/metabolism , Neurons/metabolism , Peptides/metabolism , Peptides/pharmacology , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/metabolism , Rabies virus , Structure-Activity Relationship , Viral Proteins/metabolism , Viral Proteins/pharmacology
5.
Glia ; 66(8): 1709-1723, 2018 08.
Article in English | MEDLINE | ID: mdl-29624734

ABSTRACT

Neuroinflammation and iron accumulation are hallmarks of a variety of adult neurodegenerative diseases. In Sanfilippo syndrome (mucopolysaccharidosis type III, MPSIII, a pediatric neurodegenerative disease that shares some features with adult neurodegenerative diseases), the progressive accumulation of heparan sulfate oligosaccharides (HSOs) induces microglia and astrocytes to produce pro-inflammatory cytokines leading to severe neuroinflammation. The objectives of the present study were (1) to measure the local iron concentration and to assess iron metabolism in the brain of a MPSIIIB murine model and (2) to identify the brain cells involved in this accumulation. We found that iron accumulation in MPSIIIB mice primarily affected the cerebral cortex where hepcidin levels were higher than in wild-type mice, and increased with aging. This increase was correlated with low expression of ferroportin 1 (FPN1), and thus brain iron retention. Moreover, we showed in vitro that HSOs are directly responsible for the production of hepcidin and the relative decrease in FPN1 expression when added to cultures of microglia and, to a lesser extent, to cultures of astrocytes. In contrast, no significant differences were observed in neurons. Hepcidin induction results from activation of the TLR4 pathway and STAT3 signaling, and leads to iron retention within microglia. Our results show that microglia have a key role in cerebral hepcidin overexpression and thus in the brain iron accumulation observed in the MPSIIIB model.


Subject(s)
Brain/metabolism , Iron/metabolism , Microglia/metabolism , Mucopolysaccharidosis III/metabolism , Animals , Astrocytes/metabolism , Mice, Knockout , Neurodegenerative Diseases/metabolism , Neurons/metabolism
6.
Toxicol Appl Pharmacol ; 334: 18-23, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28865757

ABSTRACT

Aerosolized liposomal amphotericin B (L-AmB) has been investigated as prophylaxis against invasive aspergillosis. However, the clinical results are controversial and some trials suggest that toxicity could be a limitation for wider use. Our aim was to assess the dynamics of cell toxicity induced in a human alveolar epithelial cell line (A549) after exposure to L-AmB (50 to 400µg/ml) or amphotericin B deoxycholate (D-AmB; 50 to 200µg/ml) by monitoring real-time A549 cell viability using an impedance-based technology. Results were expressed as cell index values integrating cell adhesion, proliferation, and survival. In parallel, the gene expression of proinflammatory cytokines was quantified at 6 and 24h after drug addition by real-time RT-PCR on cell lysates. No sustained reduction of cell indexes was observed with L-AmB or empty liposomes, even at 400µg/ml. Only the highest concentration tested of L-AmB (400µg/ml) yielded transient significant 6-fold and 4-fold induction of TNF-α and IL-8 mRNAs, respectively. In contrast, D-AmB induced a decrease in cell indexes and only the 50µg/ml concentration of D-AmB was followed by cell recovery, higher concentrations leading to cell death. Significant 4-fold, 7-fold and 3-fold inductions of TNF-α, IL-8 and IL-33 mRNAs were also observed at 6h with 50µg/ml of D-AmB. In conclusion, continuous cell impedance measurement showed no toxicity on overall cellular behavior although a slight proinflammatory cytokine expression is possible after L-AmB challenge. Real-time kinetics of cell impedance is an interesting tool for initial screening of cell toxicity.


Subject(s)
Aerosols/toxicity , Amphotericin B/toxicity , Antifungal Agents/toxicity , Deoxycholic Acid/toxicity , Electric Impedance , Epithelial Cells/drug effects , A549 Cells , Amphotericin B/chemistry , Antifungal Agents/chemistry , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokines/genetics , Cytokines/metabolism , Dosage Forms , Drug Combinations , Gene Expression Regulation/drug effects , Humans
7.
Toxins (Basel) ; 9(7)2017 06 22.
Article in English | MEDLINE | ID: mdl-28640227

ABSTRACT

Exposure to molds and mycotoxins not only contributes to the onset of respiratory disease, it also affects the ocular surface. Very few published studies concern the evaluation of the effect of mycotoxin exposure on ocular cells. The present study investigates the effects of aflatoxin B1 (AFB1) and gliotoxin, two mycotoxins secreted by Aspergillus molds, on the biological activity of the human corneal epithelial (HCE) cells. After 24, 48, and 72 h of exposure, cellular viability and inflammatory response were assessed. Both endpoint cell viability colorimetric assays and continuous cell impedance measurements, providing noninvasive real-time assessment of the effect on cells, were performed. Cytokine gene expression and interleukin-8 release were quantified. Gliotoxin appeared more cytotoxic than AFB1 but, at the same time, led to a lower increase of the inflammatory response reflecting its immunosuppressive properties. Real-time cell impedance measurement showed a distinct profile of cytotoxicity for both mycotoxins. HCE cells appeared to be a well-suited in vitro model to study ocular surface reactivity following biological contaminant exposure. Low, but persistent inflammation, caused by environmental factors, such as fungal toxins, leads to irritation and sensitization, and could be responsible for allergic manifestations which, in turn, could lead to mucosal hyper-reactivity.


Subject(s)
Aflatoxin B1/toxicity , Epithelial Cells/drug effects , Gliotoxin/toxicity , Inflammation/chemically induced , Aspergillus/metabolism , Cell Line , Cell Survival/drug effects , Cornea/cytology , Cytokines/genetics , Epithelial Cells/metabolism , Gene Expression , Humans , Inflammation/genetics
8.
Cell Microbiol ; 19(8)2017 08.
Article in English | MEDLINE | ID: mdl-28296078

ABSTRACT

Botulinum neurotoxins (BoNTs) are responsible for severe flaccid paralysis by inhibiting the release of acetylcholine at the neuromuscular junctions. BoNT type B (BoNT/B) most often induces mild forms of botulism with predominant dysautonomic symptoms. In food borne botulism and botulism by intestinal colonisation such as infant botulism, which are the most frequent naturally acquired forms of botulism, the digestive tract is the main entry route of BoNTs into the organism. We previously showed that BoNT/B translocates through mouse intestinal barrier by an endocytosis-dependent mechanism and subsequently targets neuronal cells, mainly cholinergic neurons, in the intestinal mucosa and musculosa. Here, we investigated the entry pathway of BoNT/B using fluorescent C-terminal domain of the heavy chain (HcB), which is involved in the binding to specific receptor(s) and entry process into target cells. While the combination of gangliosides GD1a /GD1b /GT1b and synaptotagmin I and to a greater extent synaptotagmin II constitutes the functional HcB receptor on NG108-15 neuronal cells, HcB only uses the gangliosides GD1a /GD1b /GT1b to efficiently bind to m-ICcl2 intestinal cells. HcB enters both cell types by a dynamin-dependent endocytosis, which is efficiently prevented by Dynasore, a dynamin inhibitor, and reaches a common early endosomal compartment labeled by early endosome antigen (EEA1). In contrast to neuronal cells, HcB uses a Cdc42-dependent pathway to enter intestinal cells. Then, HcB is transported to late endosomes in neuronal cells, whereas it exploits a nonacidified pathway from apical to basal lateral side of m-ICcl2 cells supporting a transcytotic route in epithelial intestinal cells.


Subject(s)
Botulinum Toxins, Type A/metabolism , Endocytosis , Epithelial Cells/metabolism , Neurons/metabolism , cdc42 GTP-Binding Protein/metabolism , Animals , Mice
9.
J Virol ; 90(21): 9683-9692, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27535047

ABSTRACT

RNA viruses present an extraordinary threat to human health, given their sudden and unpredictable appearance, the potential for rapid spread among the human population, and their ability to evolve resistance to antiviral therapies. The recent emergence of chikungunya virus, Zika virus, and Ebola virus highlights the struggles to contain outbreaks. A significant hurdle is the availability of antivirals to treat the infected or protect at-risk populations. While several compounds show promise in vitro and in vivo, these outbreaks underscore the need to accelerate drug discovery. The replication of several viruses has been described to rely on host polyamines, small and abundant positively charged molecules found in the cell. Here, we describe the antiviral effects of two molecules that alter polyamine levels: difluoromethylornithine (DFMO; also called eflornithine), which is a suicide inhibitor of ornithine decarboxylase 1 (ODC1), and diethylnorspermine (DENSpm), an activator of spermidine/spermine N1-acetyltransferase (SAT1). We show that reducing polyamine levels has a negative effect on diverse RNA viruses, including several viruses involved in recent outbreaks, in vitro and in vivo These findings highlight the importance of the polyamine biosynthetic pathway to viral replication, as well as its potential as a target in the development of further antivirals or currently available molecules, such as DFMO. IMPORTANCE: RNA viruses present a significant hazard to human health, and combatting these viruses requires the exploration of new avenues for targeting viral replication. Polyamines, small positively charged molecules within the cell, have been demonstrated to facilitate infection for a few different viruses. Our study demonstrates that diverse RNA viruses rely on the polyamine pathway for replication and highlights polyamine biosynthesis as a promising drug target.


Subject(s)
Antiviral Agents/pharmacology , Polyamines/metabolism , RNA Viruses/drug effects , Acetyltransferases/metabolism , Animals , Cell Line , Chikungunya Fever/drug therapy , Chikungunya Fever/virology , Chikungunya virus/drug effects , Chikungunya virus/metabolism , Disease Outbreaks , Ebolavirus/drug effects , Ebolavirus/metabolism , Eflornithine/pharmacology , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , Humans , Mice , Mice, Inbred C57BL , Spermine/analogs & derivatives , Spermine/pharmacology , Virus Replication/drug effects , Zika Virus/drug effects , Zika Virus Infection/drug therapy , Zika Virus Infection/virology
10.
J Mol Biol ; 427(4): 775-791, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25268803

ABSTRACT

Mucopolysaccharidoses type III (MPSIII, Sanfilippo syndrome) are genetic diseases due to deficient heparan sulfate (HS) saccharide digestion by lysosomal exoglycanases. Progressive accumulation of undigested saccharides causes early-onset behavioural and cognitive symptoms. The precise role of these saccharides in the pathophysiological cascade is still unclear. We showed that exposure of wild-type neural cells to exogenous soluble HS fragments of at least eight saccharides activated integrin-based focal adhesions (FAs), which attach cells to the extracellular matrix. FAs were constitutively activated in MPSIII type B astrocytes or neural stem cells unless undigested saccharides were cleared by exogenous supply of the missing exoglycanase. Defective cell polarisation and oriented migration in response to focal extracellular stimuli in affected cells suggest improper sensing of the environment. We consistently observed abnormal organisation of the rostral migratory stream in the brain of adult mice with MPSIII type B. These results suggest that cell polarisation and oriented migration defects participate to the neurological disorders associated with Sanfilippo syndrome.


Subject(s)
Astrocytes/metabolism , Focal Adhesions/metabolism , Heparitin Sulfate/pharmacology , Mucopolysaccharidosis III/pathology , Neural Stem Cells/metabolism , Animals , Astrocytes/cytology , Brain/pathology , Cell Movement/genetics , Cell Polarity/genetics , Cells, Cultured , Enzyme Activation , Focal Adhesion Kinase 1/metabolism , Heparitin Sulfate/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/cytology , Paxillin/biosynthesis , Paxillin/genetics , Phosphorylation , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Swine
11.
Mol Genet Metab ; 114(2): 138-45, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25541102

ABSTRACT

Mucopolysaccharidosis type I (MPS I) is due to deficient alpha-L-iduronidase (IDUA) which leads to storage of undegraded glycosaminoglycans (GAG). The severe form of the disease is characterized by mental retardation of unknown etiology. Trying to unveil the mechanisms that lead to cognitive impairment in MPS I, we studied alterations in the proteome from MPS I mouse hippocampus. Eight-month old mice presented increased LAMP-1 expression, GAG storage in neurons and glial cells, and impaired aversive and non-aversive memory. Shotgun proteomics was performed and 297 proteins were identified. Of those, 32 were differentially expressed. We found elevation in proteins such as cathepsins B and D; however their increase did not lead to cell death in MPS I brains. Glial fibrillary acid protein (GFAP) was markedly elevated, and immunohistochemistry confirmed a neuroinflammatory process that could be responsible for neuronal dysfunction. We didn't observe any differences in ubiquitin expression, as well as in other proteins related to protein folding, suggesting that the ubiquitin system is working properly. Finally, we observed alterations in several proteins involved in synaptic plasticity, including overexpression of post synaptic density-95 (PSD95) and reduction of microtubule-associated proteins 1A and 1B. These results together suggest that the cognitive impairment in MPS I mice is not due to massive cell death, but rather to neuronal dysfunction caused by multiple processes, including neuroinflammation and alterations in synaptic plasticity.


Subject(s)
Cognition Disorders/etiology , Cognition , Hippocampus/metabolism , Mucopolysaccharidosis I/complications , Mucopolysaccharidosis I/metabolism , Proteome/analysis , Proteomics , Animals , Brain/physiopathology , Cathepsin B/metabolism , Cathepsin D/metabolism , Cathepsin D/pharmacology , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Glycosaminoglycans/metabolism , Hippocampus/physiopathology , Iduronidase/deficiency , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Mice , Mucopolysaccharidosis I/physiopathology , Neuroglia/metabolism , Neurons/metabolism
12.
Hum Mol Genet ; 21(7): 1481-95, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22156940

ABSTRACT

Cell pathology in lysosomal storage diseases is characterized by the formation of distended vacuoles with characteristics of lysosomes. Our previous studies in mucopolysaccharidosis type IIIB (MPSIIIB), a disease in which a genetic defect induces the accumulation of undigested heparan sulfate (HS) fragments, led to the hypothesis that abnormal lysosome formation was related to events occurring at the Golgi level. We reproduced the enzyme defect of MPSIIIB in HeLa cells using tetracycline-inducible expression of shRNAs directed against α-N-acetylglucosaminidase (NAGLU) and addressed this hypothesis. HeLa cells deprived of NAGLU accumulated abnormal lysosomes. The Golgi matrix protein GM130 was over-expressed. The cis- and medial-Golgi compartments were distended, elongated and formed circularized ribbons. The Golgi microtubule network was enlarged with increased amounts of AKAP450, a partner of GM130 controlling this network. GM130 down-regulation prevented pathology in HeLa cells deprived of NAGLU, whereas GM130 over-expression in control HeLa cells mimicked the pathology of deprived cells. We concluded that abnormal lysosomes forming in cells accumulating HS fragments were the consequence of GM130 gain-of-function and subsequent alterations of the Golgi ribbon architecture. These results indicate that GM130 functions are modulated by HS glycosaminoglycans and therefore possibly controlled by extracellular cues.


Subject(s)
Autoantigens/metabolism , Membrane Proteins/metabolism , Mucopolysaccharidosis III/pathology , Acetylglucosaminidase/antagonists & inhibitors , Golgi Apparatus/ultrastructure , HeLa Cells , Humans , Lysosomes/pathology , Microtubules/ultrastructure , Models, Biological , Vacuoles/ultrastructure
13.
Hum Mol Genet ; 20(18): 3653-66, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21685203

ABSTRACT

By providing access to affected neurons, human induced pluripotent stem cells (iPSc) offer a unique opportunity to model human neurodegenerative diseases. We generated human iPSc from the skin fibroblasts of children with mucopolysaccharidosis type IIIB. In this fatal lysosomal storage disease, defective α-N-acetylglucosaminidase interrupts the degradation of heparan sulfate (HS) proteoglycans and induces cell disorders predominating in the central nervous system, causing relentless progression toward severe mental retardation. Partially digested proteoglycans, which affect fibroblast growth factor signaling, accumulated in patient cells. They impaired isolation of emerging iPSc unless exogenous supply of the missing enzyme cleared storage and restored cell proliferation. After several passages, patient iPSc starved of an exogenous enzyme continued to proliferate in the presence of fibroblast growth factor despite HS accumulation. Survival and neural differentiation of patient iPSc were comparable with unaffected controls. Whereas cell pathology was modest in floating neurosphere cultures, undifferentiated patient iPSc and their neuronal progeny expressed cell disorders consisting of storage vesicles and severe disorganization of Golgi ribbons associated with modified expression of the Golgi matrix protein GM130. Gene expression profiling in neural stem cells pointed to alterations of extracellular matrix constituents and cell-matrix interactions, whereas genes associated with lysosome or Golgi apparatus functions were downregulated. Taken together, these results suggest defective responses of patient undifferentiated stem cells and neurons to environmental cues, which possibly affect Golgi organization, cell migration and neuritogenesis. This could have potential consequences on post-natal neurological development, once HS proteoglycan accumulation becomes prominent in the affected child brain.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Lysosomes/metabolism , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/physiopathology , Neurons/cytology , Acetylglucosaminidase/genetics , Acetylglucosaminidase/metabolism , Cell Proliferation , Cells, Cultured , Child , Child, Preschool , Female , Fibroblasts/cytology , Fibroblasts/enzymology , Fibroblasts/metabolism , Heparan Sulfate Proteoglycans/metabolism , Humans , Induced Pluripotent Stem Cells/enzymology , Induced Pluripotent Stem Cells/metabolism , Lysosomes/enzymology , Male , Models, Biological , Mucopolysaccharidosis III/enzymology , Mucopolysaccharidosis III/genetics , Mutation , Neurons/enzymology , Neurons/metabolism
14.
Neurosurgery ; 68(2): 450-61; discussion 461, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21135740

ABSTRACT

BACKGROUND: Spinal root avulsion, or section, results in devastating functional sequels. Whereas reconstruction of motor pathways based on neurotization can reduce motor deficit, associated permanent limb anesthesia limits expected benefit. Sensory pathway reconstruction after dorsal root injury is limited by the inability of re-growing central sensory axons to enter the spinal cord through an injured root. OBJECTIVE: To provide evidence for the reconnection of C7 DRG neurons with the central nervous system (CNS) after experimental section of the C7 dorsal root in adult rats. METHODS: We assessed a new reconstruction strategy in adult rats 9 weeks after transection of C6 and C7 dorsal roots. Re-growing C7 central sensory axons were redirected to the noninjured C5 dorsal root through a nerve graft by end-to-side anastomosis that did not alter the C5 conduction properties. In a subgroup of rats, surgical reconstruction was combined with lentivirus-mediated gene transfer to the nerve graft in order to overexpress neurotrophin 3 (NT-3), a neurotrophic factor that stimulates sensory axon regeneration. RESULTS: Four months after reconstruction, recording of sensory evoked potentials and fluorescent tracer transport showed electrical and physical reconnection of the C7 dorsal root ganglion neurons to the spinal cord through the reconstructed pathway. Sensory perception recovery predominated on proprioception. Axonal regrowth and perception were improved when the nerve graft overexpressed neurotrophin-3 at the time of transplantation. Neurotrophin-3 overexpression did not persist 4 months after transplantation. CONCLUSION: Efficient and functional reconnection of dorsal root ganglion neurons to the spinal cord can be achieved in rats several weeks after cervical dorsal root injury. Surgical repair of sensory pathways could be considered in combination with motor nerve neurotization to treat persisting severe upper limb disability in humans.


Subject(s)
Nerve Regeneration/physiology , Neurotrophin 3/metabolism , Spinal Cord Injuries/surgery , Spinal Nerve Roots/surgery , Anastomosis, Surgical , Animals , Axotomy , Cervical Vertebrae , Evoked Potentials, Somatosensory , Male , Peroneal Nerve/transplantation , Rats , Rats, Inbred F344 , Spinal Nerve Roots/injuries , Transplants
15.
Biochem Soc Trans ; 38(6): 1442-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21118104

ABSTRACT

Biochemical disorders in lysosomal storage diseases consist of the interruption of metabolic pathways involved in the recycling of the degradation products of one or several types of macromolecules. The progressive accumulation of these primary storage products is the direct consequence of the genetic defect and represents the initial pathogenic event. Downstream consequences for the affected cells include the accumulation of secondary storage products and the formation of histological storage lesions, which appear as intracellular vacuoles that represent the pathological hallmark of lysosomal storage diseases. Relationships between storage products and storage lesions are not simple and are still largely not understood. Primary storage products induce malfunction of the organelles where they accumulate, these being primarily, but not only, lysosomes. Consequences for cell metabolism and intracellular trafficking combine the effects of primary storage product toxicity and the compensatory mechanisms activated to protect the cell. Induced disorders extend far beyond the primarily interrupted metabolic pathway.


Subject(s)
Lysosomal Storage Diseases/metabolism , Lysosomes/metabolism , Metabolic Networks and Pathways , Animals , Autoantigens/metabolism , Biological Transport , Brain/metabolism , Brain/pathology , Golgi Apparatus/pathology , Humans , Lysosomal Storage Diseases/pathology , Lysosomes/pathology , Membrane Proteins/metabolism
16.
Am J Pathol ; 177(6): 2984-99, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21037080

ABSTRACT

The accumulation of intracellular storage vesicles is a hallmark of lysosomal storage diseases. Neither the identity nor origin of these implicated storage vesicles have yet been established. The vesicles are often considered as lysosomes, endosomes, and/or autophagosomes that are engorged with undigested materials. Our studies in the mouse model of mucopolysaccharidosis type IIIB, a lysosomal storage disease that induces neurodegeneration, showed that large storage vesicles in cortical neurons did not receive material from either the endocytic or autophagy pathway, which functioned normally. Storage vesicles expressed GM130, a Golgi matrix protein, which mediates vesicle tethering in both pre- and cis-Golgi compartments. However, other components of the tethering/fusion complex were not associated with GM130 on storage vesicles, likely accounting for both the resistance of the vesicles to brefeldin A and the alteration of Golgi ribbon architecture, which comprised distended cisterna connected to LAMP1-positive storage vesicles. We propose that alteration in the GM130-mediated control of vesicle trafficking in pre-Golgi and Golgi compartments affects Golgi biogenesis and gives rise to a dead-end storage compartment. Vesicle accumulation, Golgi disorganization, and alterations of other GM130 functions may account for neuron dysfunction and death.


Subject(s)
Cytoplasmic Vesicles/pathology , Golgi Apparatus/pathology , Neurons/ultrastructure , Animals , Autophagy/physiology , Biological Transport/physiology , Cells, Cultured , Cytoplasmic Vesicles/metabolism , Disease Models, Animal , Golgi Apparatus/metabolism , Lysosomes/metabolism , Lysosomes/pathology , Membrane Proteins/metabolism , Metabolic Networks and Pathways/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucopolysaccharidosis III/complications , Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/pathology , Neurons/metabolism , Neurons/pathology , Neurons/physiology
17.
J Neurosci Res ; 88(1): 202-13, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19658197

ABSTRACT

Behavioral manifestations mark the onset of disease expression in children with mucopolysaccharidosis type III (MPSIII, Sanfilippo syndrome), a genetic disorder resulting from interruption of the lysosomal degradation of heparan sulfate. In the mouse model of MPSIII type B (MPSIIIB), cortical neuron pathology and dysfunction occur several months before neuronal loss and are primarily cell autonomous. The gene coding for GAP43, a neurite growth potentiator, is overexpressed in the MPSIIIB mouse cortex, and neurite dystrophy was reported in other types of lysosomal storage diseases. We therefore examined the development of the neuritic trees in pure populations of MPSIIIB mouse embryo cortical neurons grown for up to 12 days in primary culture. Dynamic observation of living neurons and quantification of neurite growth parameters indicated more frequent neurite elongation and branching and less frequent neurite retraction, resulting in a relative overgrowth of MPSIIIB neuron neuritic trees, involving both dendrites and axons, compared with normal controls. Neurite overgrowth was concomitant with more than twofold increased expression of GAP43 mRNAs and proteins. Correction of the genetic defect leads to expression of the missing lysosomal enzyme, normal GAP43 mRNA expression, and reduced neurite outgrowth. These results indicate that heparan sulfate oligosaccharide storage modifies GAP43 expression in MPSIIIB cortical neurons with potential consequences for neurite development and neuronal functions that may be relevant to clinical manifestations.


Subject(s)
Cerebral Cortex/metabolism , GAP-43 Protein/metabolism , Mucopolysaccharidosis III/metabolism , Neurites/metabolism , Animals , Blotting, Western , Cell Shape/physiology , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Fluorescent Antibody Technique , GAP-43 Protein/genetics , Gene Expression , Genetic Vectors/metabolism , Lentivirus/metabolism , Mice , Mucopolysaccharidosis III/genetics , Neuronal Plasticity/physiology , Neurons/cytology , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric , Time Factors
18.
Mol Cell Neurosci ; 41(1): 8-18, 2009 May.
Article in English | MEDLINE | ID: mdl-19386237

ABSTRACT

The interruption of the lysosomal degradation of heparan sulfate oligosaccharides has deleterious consequences on the central nervous system in children or in animals with mucopolysaccharidosis type III (Sanfilippo syndrome). Behavioural manifestations are prominent at disease onset, suggesting possible early synaptic defects in cortical neurons. We report that synaptophysin, the most abundant protein of the synaptic vesicle membrane, was detected at low levels in the rostral cortex of MPSIII type B mice as early as 10 days after birth. This defect preceded other disease manifestations, was associated with normal neuron and synapse density and corrected after gene transfer inducing re-expression of the missing lysosomal enzyme. Clearance of heparan sulfate oligosaccharides in cultured embryonic MPSIIIB cortical neurons or treatment with proteasome inhibitors restored normal synaptophysin levels indicating that heparan sulfate oligosaccharides activate the degradation of synaptophysin by the proteasome with consequences on synaptic vesicle components that are relevant to clinical manifestations.


Subject(s)
Mucopolysaccharidosis III/metabolism , Mucopolysaccharidosis III/physiopathology , Proteasome Endopeptidase Complex/metabolism , Synaptophysin/metabolism , Acetylglucosaminidase/genetics , Acetylglucosaminidase/metabolism , Animals , Behavior/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Child , Female , GAP-43 Protein/metabolism , Heparitin Sulfate/metabolism , Humans , Lysosomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucopolysaccharidosis III/pathology , Neurons/cytology , Neurons/metabolism , R-SNARE Proteins/metabolism , Synaptophysin/genetics
19.
PLoS One ; 3(5): e2296, 2008 May 28.
Article in English | MEDLINE | ID: mdl-18509511

ABSTRACT

BACKGROUND: In mucopolysaccharidosis type IIIB, a lysosomal storage disease causing early onset mental retardation in children, the production of abnormal oligosaccharidic fragments of heparan sulfate is associated with severe neuropathology and chronic brain inflammation. We addressed causative links between the biochemical, pathological and inflammatory disorders in a mouse model of this disease. METHODOLOGY/PRINCIPAL FINDINGS: In cell culture, heparan sulfate oligosaccharides activated microglial cells by signaling through the Toll-like receptor 4 and the adaptor protein MyD88. CD11b positive microglial cells and three-fold increased expression of mRNAs coding for the chemokine MIP1alpha were observed at 10 days in the brain cortex of MPSIIIB mice, but not in MPSIIIB mice deleted for the expression of Toll-like receptor 4 or the adaptor protein MyD88, indicating early priming of microglial cells by heparan sulfate oligosaccharides in the MPSIIIB mouse brain. Whereas the onset of brain inflammation was delayed for several months in doubly mutant versus MPSIIIB mice, the onset of disease markers expression was unchanged, indicating similar progression of the neurodegenerative process in the absence of microglial cell priming by heparan sulfate oligosaccharides. In contrast to younger mice, inflammation in aged MPSIIIB mice was not affected by TLR4/MyD88 deficiency. CONCLUSIONS/SIGNIFICANCE: These results indicate priming of microglia by HS oligosaccharides through the TLR4/MyD88 pathway. Although intrinsic to the disease, this phenomenon is not a major determinant of the neurodegenerative process. Inflammation may still contribute to neurodegeneration in late stages of the disease, albeit independent of TLR4/MyD88. The results support the view that neurodegeneration is primarily cell autonomous in this pediatric disease.


Subject(s)
Brain/metabolism , Heparitin Sulfate/metabolism , Microglia/metabolism , Mucopolysaccharidosis III/pathology , Animals , Base Sequence , Brain/pathology , Child , DNA Primers , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gene Transfer Techniques , Humans , Mice , Mice, Mutant Strains , Mucopolysaccharidosis III/metabolism
20.
Mol Genet Metab ; 94(1): 135-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18325808

ABSTRACT

Mucopolysaccharidosis IIIB is a lysosomal disease characterized by a severe neurological deterioration, the pathophysiological mechanisms of which are poorly understood. Recently FGF pathway was shown to be altered leading us to explore a downstream target involved in brain development: the collapsin response mediator protein-1 (CRMP-1). CRMP-1 transcript level was normal but a cleavage of CRMP-1 was observed with an abnormal expression of the truncated form until adult age. This truncated CRMP-1 protein could play a role in post-natal cortex maturation and be involved in neuronal alterations occurring in lysosomal diseases.


Subject(s)
Cerebral Cortex/metabolism , Mucopolysaccharidosis III/genetics , Nerve Tissue Proteins/genetics , Phosphoproteins/genetics , Animals , Calpain/metabolism , Fibroblast Growth Factors/metabolism , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mucopolysaccharidosis III/metabolism , Nerve Tissue Proteins/metabolism , Phosphoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...