Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Mol Med (Berl) ; 100(8): 1169-1179, 2022 08.
Article in English | MEDLINE | ID: mdl-35816218

ABSTRACT

Mucopolysaccharidosis type II (MPS II) is a neurometabolic disorder, due to the deficit of the lysosomal hydrolase iduronate 2-sulfatase (IDS). This leads to a severe clinical condition caused by a multi-organ accumulation of the glycosaminoglycans (GAGs/GAG) heparan- and dermatan-sulfate, whose elevated levels can be detected in body fluids. Since 2006, enzyme replacement therapy (ERT) has been clinically applied, showing efficacy in some peripheral districts. In addition to clinical monitoring, GAG dosage has been commonly used to evaluate ERT efficacy. However, a strict long-term monitoring of GAG content and composition in body fluids has been rarely performed. Here, we report the characterization of plasma and urine GAGs in Ids knock-out (Ids-ko) compared to wild-type (WT) mice, and their changes along a 24-week follow-up, with and without ERT. The concentration of heparan-sulfate (HS), chondroitin-sulfate (CS), and dermatan-sulfate (DS), and of the non-sulfated hyaluronic acid (HA), together with their differentially sulfated species, was quantified by capillary electrophoresis with laser-induced fluorescence. In untreated Ids-ko mice, HS and CS + DS were noticeably increased at all time points, while during ERT follow-up, a substantial decrease was evidenced for HS and, to a minor extent, for CS + DS. Moreover, several structural parameters were altered in untreated ko mice and reduced after ERT, however without reaching physiological values. Among these, disaccharide B and HS 2s disaccharide showed to be the most interesting candidates as biomarkers for MPS II. GAG chemical signature here defined provides potential biomarkers useful for an early diagnosis of MPS II, a more accurate follow-up of ERT, and efficacy evaluations of newly proposed therapies. KEY MESSAGES : Plasmatic and urinary GAGs are useful markers for MPS II early diagnosis and prognosis. CE-LIF allows GAG structural analysis and the quantification of 17 different disaccharides. Most GAG species increase and many structural features are altered in MPS II mouse model. GAG alterations tend to restore to wild-type levels following ERT administration. CS+DS/HS ratio, % 2,4dis CS+DS, and % HS 2s are potential markers for MPS II pathology and ERT efficacy.


Subject(s)
Body Fluids , Mucopolysaccharidosis II , Animals , Biomarkers , Body Fluids/chemistry , Dermatan Sulfate/therapeutic use , Disaccharides/analysis , Disaccharides/therapeutic use , Disease Models, Animal , Enzyme Replacement Therapy , Glycosaminoglycans , Heparitin Sulfate/therapeutic use , Mice , Mice, Knockout , Mucopolysaccharidosis II/diagnosis , Mucopolysaccharidosis II/drug therapy
2.
Biomedicines ; 9(3)2021 Mar 07.
Article in English | MEDLINE | ID: mdl-33800050

ABSTRACT

Lysosomal storage disorders (LSDs) represent a complex and heterogeneous group of rare genetic diseases due to mutations in genes coding for lysosomal enzymes, membrane proteins or transporters. This leads to the accumulation of undegraded materials within lysosomes and a broad range of severe clinical features, often including the impairment of central nervous system (CNS). When available, enzyme replacement therapy slows the disease progression although it is not curative; also, most recombinant enzymes cannot cross the blood-brain barrier, leaving the CNS untreated. The inefficient degradative capability of the lysosomes has a negative impact on the flux through the endolysosomal and autophagic pathways; therefore, dysregulation of these pathways is increasingly emerging as a relevant disease mechanism in LSDs. In the last twenty years, different LSD Drosophila models have been generated, mainly for diseases presenting with neurological involvement. The fruit fly provides a large selection of tools to investigate lysosomes, autophagy and endocytic pathways in vivo, as well as to analyse neuronal and glial cells. The possibility to use Drosophila in drug repurposing and discovery makes it an attractive model for LSDs lacking effective therapies. Here, ee describe the major cellular pathways implicated in LSDs pathogenesis, the approaches available for their study and the Drosophila models developed for these diseases. Finally, we highlight a possible use of LSDs Drosophila models for drug screening studies.

3.
Cells ; 11(1)2021 12 31.
Article in English | MEDLINE | ID: mdl-35011691

ABSTRACT

Deficit of the IDUA (α-L-iduronidase) enzyme causes the lysosomal storage disorder mucopolysaccharidosis type I (MPS I), a rare pediatric neurometabolic disease, due to pathological variants in the IDUA gene and is characterized by the accumulation of the undegraded mucopolysaccharides heparan sulfate and dermatan sulfate into lysosomes, with secondary cellular consequences that are still mostly unclarified. Here, we report a new fruit fly RNAi-mediated knockdown model of a IDUA homolog (D-idua) displaying a phenotype mimicking some typical molecular features of Lysosomal Storage Disorders (LSD). In this study, we showed that D-idua is a vital gene in Drosophila and that ubiquitous reduction of its expression leads to lethality during the pupal stage, when the precise degradation/synthesis of macromolecules, together with a functional autophagic pathway, are indispensable for the correct development to the adult stage. Tissue-specific analysis of the D-idua model showed an increase in the number and size of lysosomes in the brain and muscle. Moreover, the incorrect acidification of lysosomes led to dysfunctional lysosome-autophagosome fusion and the consequent block of autophagy flux. A concomitant metabolic drift of glycolysis and lipogenesis pathways was observed. After starvation, D-idua larvae showed a quite complete rescue of both autophagy/lysosome phenotypes and metabolic alterations. Metabolism and autophagy are strictly interconnected vital processes that contribute to maintain homeostatic control of energy balance, and little is known about this regulation in LSDs. Our results provide new starting points for future investigations on the disease's pathogenic mechanisms and possible pharmacological manipulations.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Mucopolysaccharidosis I/enzymology , Mucopolysaccharidosis I/pathology , Amino Acid Sequence , Animals , Autophagosomes/metabolism , Autophagy , Disease Models, Animal , Down-Regulation/genetics , Drosophila Proteins/chemistry , Drosophila melanogaster/genetics , Genes, Essential , Glycolysis , Lipogenesis , Locomotion , Longevity , Lysosomes/metabolism , Muscles/metabolism , Organ Specificity , Phenotype , RNA Interference
4.
Life (Basel) ; 10(11)2020 Oct 30.
Article in English | MEDLINE | ID: mdl-33142967

ABSTRACT

Mucopolysaccharidosis type II (MPS II) is a lysosomal storage disorder that occurs due to the deficit of the lysosomal enzyme iduronate 2-sulfatase (IDS) that leads to the storage of the glycosaminoglycan heparan- and dermatan-sulfate in all organs and tissues. It is characterized by important clinical features and the severe form presents with a heavy neurological involvement. However, almost nothing is known about the neuropathogenesis of MPS II. To address this issue, we developed a ubiquitous, neuronal, and glial-specific knockdown model in Drosophila melanogaster by using the RNA interference (RNAi) approach. Knockdown of the Ids/CG12014 gene resulted in a significant reduction of the Ids gene expression and enzymatic activity. However, glycosaminoglycan storage, survival, molecular markers (Atg8a, Lamp1, Rab11), and locomotion behavior were not affected. Even strongly reduced, IDS-activity was enough to prevent a pathological phenotype in a MPS II RNAi fruit fly. Thus, a Drosophila MPS II model requires complete abolishment of the enzymatic activity.

5.
Int J Mol Sci ; 21(4)2020 Feb 13.
Article in English | MEDLINE | ID: mdl-32070051

ABSTRACT

Mucopolysaccharidosis type II (MPS II, Hunter syndrome) was first described by Dr. Charles Hunter in 1917. Since then, about one hundred years have passed and Hunter syndrome, although at first neglected for a few decades and afterwards mistaken for a long time for the similar disorder Hurler syndrome, has been clearly distinguished as a specific disease since 1978, when the distinct genetic causes of the two disorders were finally identified. MPS II is a rare genetic disorder, recently described as presenting an incidence rate ranging from 0.38 to 1.09 per 100,000 live male births, and it is the only X-linked-inherited mucopolysaccharidosis. The complex disease is due to a deficit of the lysosomal hydrolase iduronate 2-sulphatase, which is a crucial enzyme in the stepwise degradation of heparan and dermatan sulphate. This contributes to a heavy clinical phenotype involving most organ-systems, including the brain, in at least two-thirds of cases. In this review, we will summarize the history of the disease during this century through clinical and laboratory evaluations that allowed its definition, its correct diagnosis, a partial comprehension of its pathogenesis, and the proposition of therapeutic protocols. We will also highlight the main open issues related to the possible inclusion of MPS II in newborn screenings, the comprehension of brain pathogenesis, and treatment of the neurological compartment.


Subject(s)
Genes, X-Linked/genetics , Iduronate Sulfatase/genetics , Mucopolysaccharidosis II/genetics , Mucopolysaccharidosis II/therapy , Brain/pathology , Humans , Male , Mucopolysaccharidosis II/diagnosis , Mucopolysaccharidosis II/pathology , Phenotype
6.
Int J Mol Sci ; 20(8)2019 Apr 24.
Article in English | MEDLINE | ID: mdl-31022913

ABSTRACT

Mucopolysaccharidosis type II (MPSII) is a lysosomal storage disorder due to the deficit of the enzyme iduronate 2-sulfatase (IDS), which leads to the accumulation of glycosaminoglycans in most organ-systems, including the brain, and resulting in neurological involvement in about two-thirds of the patients. The main treatment is represented by a weekly infusion of the functional enzyme, which cannot cross the blood-brain barrier and reach the central nervous system. In this study, a tailored nanomedicine approach based on brain-targeted polymeric nanoparticles (g7-NPs), loaded with the therapeutic enzyme, was exploited. Fibroblasts from MPSII patients were treated for 7 days with NPs loaded with the IDS enzyme; an induced IDS activity like the one detected in healthy cells was measured, together with a reduction of GAG content to non-pathological levels. An in vivo short-term study in MPSII mice was performed by weekly administration of g7-NPs-IDS. Biochemical, histological, and immunohistochemical evaluations of liver and brain were performed. The 6-weeks treatment produced a significant reduction of GAG deposits in liver and brain tissues, as well as a reduction of some neurological and inflammatory markers (i.e., LAMP2, CD68, GFAP), highlighting a general improvement of the brain pathology. The g7-NPs-IDS approach allowed a brain-targeted enzyme replacement therapy. Based on these positive results, the future aim will be to optimize NP formulation further to gain a higher efficacy of the proposed approach.


Subject(s)
Brain/drug effects , Drug Carriers/metabolism , Drug Delivery Systems , Iduronate Sulfatase/administration & dosage , Mucopolysaccharidosis II/drug therapy , Nanoparticles/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/metabolism , Animals , Brain/enzymology , Brain/metabolism , Brain/pathology , Drug Carriers/chemistry , Enzyme Replacement Therapy , Glycopeptides/chemistry , Glycopeptides/metabolism , Humans , Iduronate Sulfatase/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mucopolysaccharidosis II/enzymology , Mucopolysaccharidosis II/metabolism , Mucopolysaccharidosis II/pathology , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry
7.
Eur J Pediatr ; 178(5): 739-753, 2019 May.
Article in English | MEDLINE | ID: mdl-30809705

ABSTRACT

Mucopolysaccharidoses (MPS) are a subgroup of 11 monogenic lysosomal storage disorders due to the deficit of activity of the lysosomal hydrolases deputed to the degradation of mucopolysaccharides. Although individually rare, all together they account for at least 1:25,000 live births. In this study, we present the genetic analysis of a population of 71 MPS patients enrolled in a multicenter Italian study. We re-annotated all variants, according to the latest recommendations, and re-classified them as suggested by the American College of Medical Genetics and Genomics. Variant distribution per type was mainly represented by missense mutations. Overall, 10 patients had received no molecular diagnosis, although 6 of them had undergone either HSCT or ERT, based on clinical and enzymatic evaluations. Moreover, nine novel variants are reported.Conclusions: Our analysis underlines the need to complete the molecular diagnosis in patients previously diagnosed only on a biochemical basis, suggests a periodical re-annotation of the variants and solicits their deposition in public databases freely available to clinicians and researchers. We strongly recommend a molecular diagnosis based on the analysis of the "trio" instead of the sole proband. These recommendations will help to obtain a complete and correct diagnosis of mucopolysaccharidosis, rendering also possible genetic counseling. What is known • MPS are a group of 11 metabolic genetic disorders due to deficits of enzymes involved in the mucopolysaccharides degradation. • Molecular analysis is commonly performed to confirm enzymatic assays. What is new • Eighty-six percent of the 71 patients we collected received a molecular diagnosis; among them, 9 novel variants were reported. • We stress the importance of molecular diagnosis in biochemically diagnosed patients, encourage a periodical re-annotation of variants according to the recent nomenclature and their publication in open databases.


Subject(s)
Genetic Testing , Molecular Diagnostic Techniques , Mucopolysaccharidoses/diagnosis , Adolescent , Adult , Child , Child, Preschool , Female , Genetic Association Studies , Genetic Counseling , Genetic Markers , Humans , Infant , Italy , Male , Mucopolysaccharidoses/genetics , Mutation, Missense , Young Adult
8.
Anal Biochem ; 557: 34-41, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30009765

ABSTRACT

Dried blood spot (DBS) technology is a cheap and easy method largely applied in newborn screening. Mucopolysaccharidoses (MPS) are characterized by the deficit of enzymes that degrade glycosaminoglycans (GAGs) characterized by progressive worsening of the conditions. For a possible early diagnosis of MPS, we developed a method of uronic acid (UA)-GAGs determination in DBS of 600 healthy newborns and from a small group of MPS subjects matched for age. Spotted blood UA-GAGs of the normal newborns are composed of 67.2% chondroitin sulfate (CS), 28.6% heparan sulfate (HS) and 4.4% hyaluronic acid with a CS/HS ratio of 2.35 and a total GAGs content of 0.43 µg/DBS. A chemical evaluation of CS and HS structure was performed by measuring their disaccharide composition, sulfation and the overall charge density. The DBS of four different MPS types presented an increase of total or single UA-GAGs content and/or modifications of the CS and HS disaccharide composition as well as chemical signature also related to the MPS enzymatic defect. The modifications of the UA-GAGs composition, parameters and structure of healthy newborns determined in DBS would be useful for a possible early diagnosis of various MPS types.


Subject(s)
Dried Blood Spot Testing , Glycosaminoglycans/blood , Glycosaminoglycans/chemistry , Mucopolysaccharidoses/blood , Mucopolysaccharidoses/diagnosis , Carbohydrate Conformation , Humans , Infant, Newborn
10.
Hum Mol Genet ; 27(13): 2262-2275, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29648648

ABSTRACT

Skeletal abnormalities represent a major clinical burden in patients affected by the lysosomal storage disorder mucopolysaccharidosis type II (MPSII, OMIM #309900). While extensive research has emphasized the detrimental role of stored glycosaminoglycans (GAGs) in the bone marrow (BM), a limited understanding of primary cellular mechanisms underlying bone defects in MPSII has hampered the development of bone-targeted therapeutic strategies beyond enzyme replacement therapy (ERT). We here investigated the involvement of key signaling pathways related to the loss of iduronate-2-sulfatase activity in two different MPSII animal models, D. rerio and M. musculus. We found that FGF pathway activity is impaired during early stages of bone development in IDS knockout mice and in a newly generated Ids mutant fish. In both models the FGF signaling deregulation anticipated a slow but progressive defect in bone differentiation, regardless of any extensive GAGs storage. We also show that MPSII patient fibroblasts harboring different mutations spanning the IDS gene exhibit perturbed FGF signaling-related markers expression. Our work opens a new venue to discover possible druggable novel key targets in MPSII.


Subject(s)
Brain/metabolism , Fibroblast Growth Factors/genetics , Iduronate Sulfatase/genetics , Mucopolysaccharidosis II/genetics , Animals , Brain/pathology , Disease Models, Animal , Enzyme Replacement Therapy , Gene Expression Regulation , Glycosaminoglycans/genetics , Humans , Iduronate Sulfatase/therapeutic use , Mice , Mice, Knockout , Mucopolysaccharidosis II/pathology , Signal Transduction , Zebrafish/genetics
11.
Int J Mol Sci ; 18(5)2017 May 17.
Article in English | MEDLINE | ID: mdl-28513549

ABSTRACT

Lysosomal storage disorders (LSDs) are a group of about 50 genetic metabolic disorders, mainly affecting children, sharing the inability to degrade specific endolysosomal substrates. This results in failure of cellular functions in many organs, including brain that in most patients may go through progressive neurodegeneration. In this study, we analyzed the brain of the mouse model for Hunter syndrome, a LSD mostly presenting with neurological involvement. Whole transcriptome analysis of the cerebral cortex and midbrain/diencephalon/hippocampus areas was performed through RNA-seq. Genes known to be involved in several neurological functions showed a significant differential expression in the animal model for the disease compared to wild type. Among the pathways altered in both areas, axon guidance, calcium homeostasis, synapse and neuroactive ligand-receptor interaction, circadian rhythm, neuroinflammation and Wnt signaling were the most significant. Application of RNA sequencing to dissect pathogenic alterations of complex syndromes allows to photograph perturbations, both determining and determined by these disorders, which could simultaneously occur in several metabolic and biochemical pathways. Results also emphasize the common, altered pathways between neurodegenerative disorders affecting elderly and those associated with pediatric diseases of genetic origin, perhaps pointing out a general common course for neurodegeneration, independent from the primary triggering cause.


Subject(s)
Brain/metabolism , Gene Expression Profiling , Mucopolysaccharidosis II/genetics , Sequence Analysis, RNA , Animals , Computational Biology/methods , Disease Models, Animal , Gene Expression Regulation , Gene Ontology , Mice , Molecular Sequence Annotation , Mucopolysaccharidosis II/metabolism , Signal Transduction , Transcriptome
13.
PLoS One ; 11(5): e0156452, 2016.
Article in English | MEDLINE | ID: mdl-27228099

ABSTRACT

Lysosomal Storage Disorders (LSDs) are a group of metabolic syndromes, each one due to the deficit of one lysosomal enzyme. Many LSDs affect most of the organ systems and overall about 75% of the patients present neurological impairment. Enzyme Replacement Therapy, although determining some systemic clinical improvements, is ineffective on the CNS disease, due to enzymes' inability to cross the blood-brain barrier (BBB). With the aim to deliver the therapeutic enzymes across the BBB, we here assayed biodegradable and biocompatible PLGA-nanoparticles (NPs) in two murine models for LSDs, Mucopolysaccharidosis type I and II (MPS I and MPS II). PLGA-NPs were modified with a 7-aminoacid glycopeptide (g7), yet demonstrated to be able to deliver low molecular weight (MW) molecules across the BBB in rodents. We specifically investigated, for the first time, the g7-NPs ability to transfer a model drug (FITC-albumin) with a high MW, comparable to the enzymes to be delivered for LSDs brain therapy. In vivo experiments, conducted on wild-type mice and knockout mouse models for MPS I and II, also included a whole series of control injections to obtain a broad preliminary view of the procedure efficiency. Results clearly showed efficient BBB crossing of albumin in all injected mice, underlying the ability of NPs to deliver high MW molecules to the brain. These results encourage successful experiments with enzyme-loaded g7-NPs to deliver sufficient amounts of the drug to the brain district on LSDs, where exerting a corrective effect on the pathological phenotype.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Carriers , Enzyme Replacement Therapy/methods , Mucopolysaccharidosis II/drug therapy , Mucopolysaccharidosis I/drug therapy , Nanoparticles , Albumins/chemistry , Albumins/pharmacokinetics , Albumins/pharmacology , Animals , Disease Models, Animal , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Fluorescein-5-isothiocyanate/chemistry , Fluorescein-5-isothiocyanate/pharmacokinetics , Fluorescein-5-isothiocyanate/pharmacology , Lactic Acid/chemistry , Lactic Acid/pharmacokinetics , Lactic Acid/pharmacology , Mice , Mice, Knockout , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis I/pathology , Mucopolysaccharidosis II/genetics , Mucopolysaccharidosis II/metabolism , Mucopolysaccharidosis II/pathology , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacokinetics , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer
14.
Eur J Oncol Nurs ; 20: 58-63, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26163026

ABSTRACT

PURPOSE: to develop a tool for measuring the difficulty of intravenous line insertion in cancer patients (DIVA-CP) receiving peripheral chemotherapy. METHODS: a pilot-validation study divided into two phases was performed in a north-eastern Italian outpatient chemotherapy centre. In the first phase, a review of the literature and brainstorming sessions/direct discussions among expert oncology nurses were used to develop items on the DIVA-CP tool, and a panel of expert oncology nurses assessed the tool face and content validity. In the second phase, 260 adult patients undergoing single chemotherapy cycles were consecutively enrolled. Data was analysed for construct validity (explorative factor analysis) and inter-rater reliability (Cohen's Kappa). RESULTS: a 10-item tool was developed with four factors that were identified through factor analysis, explaining a total variance of 61.578%: accessibility to first choice veins (23.057%), venous fragility (15.197%), probable difficulties during the procedure (12.642%), and repeated exposure (10.691%). The tool demonstrated excellent inter-rater reliability (κ > 0.61 for 7 out of 10 items). CONCLUSIONS: the DIVA-CP tool is still a pilot instrument that needs to be improved through future studies. The tool has great potential and may perform an important role in oncological settings, allowing for easier venous assessment of patients receiving peripheral chemotherapy who are at risk of difficult intravenous access insertion. In addition, this instrument may help nurses to identify patients that may benefit from a central catheter promptly.


Subject(s)
Antineoplastic Agents/administration & dosage , Catheterization, Peripheral/methods , Catheterization, Peripheral/standards , Infusions, Intravenous/methods , Infusions, Intravenous/standards , Neoplasms/drug therapy , Psychometrics/instrumentation , Factor Analysis, Statistical , Female , Humans , Italy , Male , Middle Aged , Pilot Projects , Reproducibility of Results , Surveys and Questionnaires
15.
BMC Med Genomics ; 6: 37, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-24083598

ABSTRACT

BACKGROUND: Hunter syndrome (HS) is a lysosomal storage disease caused by iduronate-2-sulfatase (IDS) deficiency and loss of ability to break down and recycle the glycosaminoglycans, heparan and dermatan sulfate, leading to impairment of cellular processes and cell death. Cell activities and functioning of intracellular organelles are controlled by the clock genes (CGs), driving the rhythmic expression of clock controlled genes (CCGs). We aimed to evaluate the expression of CGs and downstream CCGs in HS, before and after enzyme replacement treatment with IDS. METHODS: The expression levels of CGs and CCGs were evaluated by a whole transcriptome analysis through Next Generation Sequencing in normal primary human fibroblasts and fibroblasts of patients affected by HS before and 24 h/144 h after IDS treatment. The time related expression of CGs after synchronization by serum shock was also evaluated by qRT-PCR before and after 24 hours of IDS treatment. RESULTS: In HS fibroblasts we found altered expression of several CGs and CCGs, with dynamic changes 24 h and 144 h after IDS treatment. A semantic hypergraph-based analysis highlighted five gene clusters significantly associated to important biological processes or pathways, and five genes, AHR, HIF1A, CRY1, ITGA5 and EIF2B3, proven to be central players in these pathways. After synchronization by serum shock and 24 h treatment with IDS the expression of ARNTL2 at 10 h (p = 0.036), PER1 at 4 h (p = 0.019), PER2 at 10 h (p = 0.041) and 16 h (p = 0.043) changed in HS fibroblasts. CONCLUSION: CG and CCG expression is altered in HS fibroblasts and IDS treatment determines dynamic modifications, suggesting a direct involvement of the CG machinery in the physiopathology of cellular derangements that characterize HS.


Subject(s)
Circadian Rhythm/drug effects , Circadian Rhythm/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Iduronate Sulfatase/pharmacology , Mucopolysaccharidosis II/pathology , Case-Control Studies , Child , Computer Graphics , Humans , Mucopolysaccharidosis II/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Epilepsy Res Treat ; 2011: 258365, 2011.
Article in English | MEDLINE | ID: mdl-22937229

ABSTRACT

Autosomal dominant lateral temporal epilepsy (ADTLE) is an inherited epileptic syndrome characterized by ictal auditory symptoms or aphasia, negative MRI findings, and relatively benign evolution. Mutations responsible for ADLTE have been found in the LGI1 gene. The functions of the Lgi1 protein apparently are mediated by interactions with members of the ADAM protein family: it binds the postsynaptic receptor ADAM22 to regulate glutamate-AMPA currents at excitatory synapses and also the ADAM23 receptor to promote neurite outgrowth in vitro and dendritic arborization in vivo. Because alteration of each of these neuronal mechanisms may underlie ADLTE, ADAM22 and ADAM23 are candidate genes for this syndrome. In a previous work, we excluded a major role of ADAM22 in the aetiology of ADLTE. Here, we performed linkage analysis between microsatellite markers within or flanking the ADAM23 gene and ADLTE in 13 Italian families. The results exclude ADAM23 as major causative gene for ADLTE.

SELECTION OF CITATIONS
SEARCH DETAIL
...