Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 187
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 137(2): 235-46, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19379691

RESUMEN

X-linked myopathy with excessive autophagy (XMEA) is a childhood-onset disease characterized by progressive vacuolation and atrophy of skeletal muscle. We show that XMEA is caused by hypomorphic alleles of the VMA21 gene, that VMA21 is the diverged human ortholog of the yeast Vma21p protein, and that like Vma21p it is an essential assembly chaperone of the V-ATPase, the principal mammalian proton pump complex. Decreased VMA21 raises lysosomal pH, which reduces lysosomal degradative ability and blocks autophagy. This reduces cellular free amino acids, which upregulates the mTOR pathway and mTOR-dependent macroautophagy, resulting in proliferation of large and ineffective autolysosomes that engulf sections of cytoplasm, merge together, and vacuolate the cell. Our results uncover macroautophagic overcompensation leading to cell vacuolation and tissue atrophy as a mechanism of disease.


Asunto(s)
Genes Ligados a X , Enfermedades Musculares/genética , ATPasas de Translocación de Protón Vacuolares/metabolismo , Autofagia , Humanos , Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , ARN Mensajero/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , ATPasas de Translocación de Protón Vacuolares/genética
2.
Hum Genet ; 142(7): 909-925, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37183190

RESUMEN

Contactin-associated protein-like 2 (CNTNAP2) gene encodes for CASPR2, a presynaptic type 1 transmembrane protein, involved in cell-cell adhesion and synaptic interactions. Biallelic CNTNAP2 loss has been associated with "Pitt-Hopkins-like syndrome-1" (MIM#610042), while the pathogenic role of heterozygous variants remains controversial. We report 22 novel patients harboring mono- (n = 2) and bi-allelic (n = 20) CNTNAP2 variants and carried out a literature review to characterize the genotype-phenotype correlation. Patients (M:F 14:8) were aged between 3 and 19 years and affected by global developmental delay (GDD) (n = 21), moderate to profound intellectual disability (n = 17) and epilepsy (n = 21). Seizures mainly started in the first two years of life (median 22.5 months). Antiseizure medications were successful in controlling the seizures in about two-thirds of the patients. Autism spectrum disorder (ASD) and/or other neuropsychiatric comorbidities were present in nine patients (40.9%). Nonspecific midline brain anomalies were noted in most patients while focal signal abnormalities in the temporal lobes were noted in three subjects. Genotype-phenotype correlation was performed by also including 50 previously published patients (15 mono- and 35 bi-allelic variants). Overall, GDD (p < 0.0001), epilepsy (p < 0.0001), hyporeflexia (p = 0.012), ASD (p = 0.009), language impairment (p = 0.020) and severe cognitive impairment (p = 0.031) were significantly associated with the presence of biallelic versus monoallelic variants. We have defined the main features associated with biallelic CNTNAP2 variants, as severe cognitive impairment, epilepsy and behavioral abnormalities. We propose CASPR2-deficiency neurodevelopmental disorder as an exclusively recessive disease while the contribution of heterozygous variants is less likely to follow an autosomal dominant inheritance pattern.


Asunto(s)
Trastorno del Espectro Autista , Epilepsia , Humanos , Niño , Trastorno del Espectro Autista/genética , Discapacidades del Desarrollo/genética , Epilepsia/genética , Estudios de Asociación Genética , Convulsiones/genética , Contactinas/genética
3.
Biochem Biophys Res Commun ; 673: 131-136, 2023 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-37385007

RESUMEN

Aromatic l-amino acid decarboxylase (AADC) deficiency is a rare autosomal recessive neurometabolic disorder caused by biallelic pathogenic variants in the DDC gene and mainly characterized by developmental delay, hypotonia, and oculogyric crises. Early diagnosis is crucial for correct patient management; however, many patients remain misdiagnosed or undiagnosed due to the rarity and clinical heterogeneity of the disorder especially in the milder forms. Here, we applied exome sequencing approach by screening 2000 paediatric patients with neurodevelopmental disorders to identify possible new AADC variants and AADC deficiency patients. We identified five distinct DDC variants in two unrelated individuals. Patient #1 harboured two compound heterozygous DDC variants: c.436-12T > C and c.435 + 24A>C and presented with psychomotor delay, tonic spasms, and hyperreactivity. Patient #2 had three homozygous AADC variants: c.1385G > A; p.Arg462Gln, c.234C > T; p.Ala78 = , and c.201 + 37A > G and presented with developmental delay and myoclonic seizures. The variants were classified as benign class I variants and therefore non-causative according to the ACMG/AMP guidelines. Since the AADC protein is a structural and functional obligate homodimer, we evaluated the possible AADC polypeptide chain combinations in the two patients and determined the effects resulting from the amino acid substitution Arg462Gln. Our patients carrying DDC variants presented clinical manifestations not precisely overlapped to the classical symptoms exhibited by the most severe AADC deficiency cases. However, screening data derived from exome sequencing in patients featuring wide-range symptoms related to neurodevelopmental disorders may help to identify AADC deficiency patients, especially when applied to larger cohorts.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos , Trastornos del Neurodesarrollo , Humanos , Niño , Secuenciación del Exoma , Descarboxilasas de Aminoácido-L-Aromático/genética , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/genética , Trastornos del Neurodesarrollo/diagnóstico , Trastornos del Neurodesarrollo/genética , Aminoácidos/genética
4.
Cerebellum ; 22(2): 206-222, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35218524

RESUMEN

Cerebellar hypoplasia and dysplasia encompass a group of clinically and genetically heterogeneous disorders frequently associated with neurodevelopmental impairment. The Neuron Navigator 2 (NAV2) gene (MIM: 607,026) encodes a member of the Neuron Navigator protein family, widely expressed within the central nervous system (CNS), and particularly abundant in the developing cerebellum. Evidence across different species supports a pivotal function of NAV2 in cytoskeletal dynamics and neurite outgrowth. Specifically, deficiency of Nav2 in mice leads to cerebellar hypoplasia with abnormal foliation due to impaired axonal outgrowth. However, little is known about the involvement of the NAV2 gene in human disease phenotypes. In this study, we identified a female affected with neurodevelopmental impairment and a complex brain and cardiac malformations in which clinical exome sequencing led to the identification of NAV2 biallelic truncating variants. Through protein expression analysis and cell migration assay in patient-derived fibroblasts, we provide evidence linking NAV2 deficiency to cellular migration deficits. In model organisms, the overall CNS histopathology of the Nav2 hypomorphic mouse revealed developmental anomalies including cerebellar hypoplasia and dysplasia, corpus callosum hypo-dysgenesis, and agenesis of the olfactory bulbs. Lastly, we show that the NAV2 ortholog in Drosophila, sickie (sick) is widely expressed in the fly brain, and sick mutants are mostly lethal with surviving escapers showing neurobehavioral phenotypes. In summary, our results unveil a novel human neurodevelopmental disorder due to genetic loss of NAV2, highlighting a critical conserved role of the NAV2 gene in brain and cerebellar development across species.


Asunto(s)
Encéfalo , Malformaciones del Sistema Nervioso , Animales , Femenino , Humanos , Ratones , Cerebelo/anomalías , Neuronas
5.
Am J Hum Genet ; 104(4): 721-730, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30929742

RESUMEN

VAMP2 encodes the vesicular SNARE protein VAMP2 (also called synaptobrevin-2). Together with its partners syntaxin-1A and synaptosomal-associated protein 25 (SNAP25), VAMP2 mediates fusion of synaptic vesicles to release neurotransmitters. VAMP2 is essential for vesicular exocytosis and activity-dependent neurotransmitter release. Here, we report five heterozygous de novo mutations in VAMP2 in unrelated individuals presenting with a neurodevelopmental disorder characterized by axial hypotonia (which had been present since birth), intellectual disability, and autistic features. In total, we identified two single-amino-acid deletions and three non-synonymous variants affecting conserved residues within the C terminus of the VAMP2 SNARE motif. Affected individuals carrying de novo non-synonymous variants involving the C-terminal region presented a more severe phenotype with additional neurological features, including central visual impairment, hyperkinetic movement disorder, and epilepsy or electroencephalography abnormalities. Reconstituted fusion involving a lipid-mixing assay indicated impairment in vesicle fusion as one of the possible associated disease mechanisms. The genetic synaptopathy caused by VAMP2 de novo mutations highlights the key roles of this gene in human brain development and function.


Asunto(s)
Discapacidad Intelectual/genética , Hipotonía Muscular/genética , Trastornos del Neurodesarrollo/genética , Neuronas/metabolismo , Sinapsis/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/genética , Adolescente , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Encéfalo/diagnóstico por imagen , Niño , Preescolar , Epilepsia/metabolismo , Exocitosis , Femenino , Heterocigoto , Humanos , Lípidos/química , Imagen por Resonancia Magnética , Masculino , Fusión de Membrana , Trastornos del Movimiento/genética , Mutación , Trastornos del Neurodesarrollo/metabolismo , Neurotransmisores/metabolismo , Fenotipo , Dominios Proteicos , Proteínas R-SNARE/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/fisiología
6.
Muscle Nerve ; 65(1): 96-104, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34687219

RESUMEN

INTRODUCTION/AIMS: Currently, there are no straightforward guidelines for the clinical and diagnostic management of hyperCKemia, a frequent and nonspecific presentation in muscle diseases. Therefore, we aimed to describe our diagnostic workflow for evaluating patients with this condition. METHODS: We selected 83 asymptomatic or minimally symptomatic patients with persistent hyperCKemia for participation in this Italian multicenter study. Patients with facial involvement and distal or congenital myopathies were excluded, as were patients with suspected inflammatory myopathies or predominant respiratory or cardiac involvement. All patients underwent a neurological examination and nerve conduction and electromyography studies. The first step of the investigation included a screening for Pompe disease. We then evaluated the patients for myotonic dystrophy type II-related CCTG expansion and excluded patients with copy number variations in the DMD gene. Subsequently, the undiagnosed patients were investigated using a target gene panel that included 20 genes associated with isolated hyperCKemia. RESULTS: Using this approach, we established a definitive diagnosis in one third of the patients. The detection rate was higher in patients with severe hyperCKemia and abnormal electromyographic findings. DISCUSSION: We have described our diagnostic workflow for isolated hyperCKemia, which is based on electrodiagnostic data, biochemical screening, and first-line genetic investigations, followed by successive targeted sequencing panels. Both clinical signs and electromyographic abnormalities are associated with increased diagnostic yields.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , Enfermedades Musculares , Creatina Quinasa , Variaciones en el Número de Copia de ADN , Electromiografía , Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , Humanos
7.
Am J Hum Genet ; 103(3): 431-439, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30100084

RESUMEN

ADP-ribosylation, the addition of poly-ADP ribose (PAR) onto proteins, is a response signal to cellular challenges, such as excitotoxicity or oxidative stress. This process is catalyzed by a group of enzymes referred to as poly(ADP-ribose) polymerases (PARPs). Because the accumulation of proteins with this modification results in cell death, its negative regulation restores cellular homeostasis: a process mediated by poly-ADP ribose glycohydrolases (PARGs) and ADP-ribosylhydrolase proteins (ARHs). Using linkage analysis and exome or genome sequencing, we identified recessive inactivating mutations in ADPRHL2 in six families. Affected individuals exhibited a pediatric-onset neurodegenerative disorder with progressive brain atrophy, developmental regression, and seizures in association with periods of stress, such as infections. Loss of the Drosophila paralog Parg showed lethality in response to oxidative challenge that was rescued by human ADPRHL2, suggesting functional conservation. Pharmacological inhibition of PARP also rescued the phenotype, suggesting the possibility of postnatal treatment for this genetic condition.

8.
Clin Genet ; 100(5): 607-614, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34296759

RESUMEN

Early infantile epileptic encephalopathy 38 (EIEE38, MIM #617020) is caused by biallelic variants in ARV1, encoding a transmembrane protein of the endoplasmic reticulum with a pivotal role in glycosylphosphatidylinositol (GPI) biosynthesis. We ascertained seven new patients from six unrelated families harboring biallelic variants in ARV1, including five novel variants. Affected individuals showed psychomotor delay, hypotonia, early onset refractory seizures followed by regression and specific neuroimaging features. Flow cytometric analysis on patient fibroblasts showed a decrease in GPI-anchored proteins on the cell surface, supporting a lower residual activity of the mutant ARV1 as compared to the wildtype. A rescue assay through the transduction of lentivirus expressing wild type ARV1 cDNA effectively rescued these alterations. This study expands the clinical and molecular spectrum of the ARV1-related encephalopathy, confirming the essential role of ARV1 in GPI biosynthesis and brain function.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Proteínas de la Membrana/deficiencia , Fenotipo , Espasmos Infantiles/diagnóstico , Espasmos Infantiles/genética , Alelos , Sustitución de Aminoácidos , Encéfalo/anomalías , Proteínas Portadoras/genética , Análisis Mutacional de ADN , Facies , Femenino , Proteínas Ligadas a GPI/biosíntesis , Estudios de Asociación Genética/métodos , Glicosilfosfatidilinositoles/metabolismo , Humanos , Imagen por Resonancia Magnética , Masculino , Proteínas de la Membrana/genética , Mutación , Linaje , Embarazo , Diagnóstico Prenatal/métodos , Espasmos Infantiles/metabolismo
9.
Clin Neuropathol ; 40(6): 310-318, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34281632

RESUMEN

AIM: Since the immune system plays a role in the pathogenesis of several muscular dystrophies, we aim to characterize several muscular inflammatory features in α- (LGMD R3) and γ-sarcoglycanopathies (LGMD R5). MATERIALS AND METHODS: We explored the expression of major histocompatibility complex class I molecules (MHCI), and we analyzed the composition of the immune infiltrates in muscle biopsies from 10 patients with LGMD R3 and 8 patients with LGMD R5, comparing the results to Duchenne muscular dystrophy patients (DMD). RESULTS: A consistent involvement of the immune response was observed in sarcoglycanopathies, although it was less evident than in DMD. LGMD R3-R5 and DMD shared an abnormal expression of MHCI, and the composition of the muscular immune cell infiltrate was comparable. CONCLUSION: These findings might serve as a rationale to fine-tune a disease-specific immunomodulatory regimen, particularly relevant in view of the rapid development of gene therapy for sarcoglycanopathies.


Asunto(s)
Distrofias Musculares , Miositis , Sarcoglicanopatías , Biopsia , Humanos , Músculo Esquelético , Sarcoglicanopatías/genética
10.
Neurobiol Dis ; 137: 104757, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31978608

RESUMEN

Elastin microfibril interface-located proteins (EMILINs) are extracellular matrix glycoproteins implicated in elastogenesis and cell proliferation. Recently, a missense mutation in the EMILIN1 gene has been associated with autosomal dominant connective tissue disorder and motor-sensory neuropathy in a single family. We identified by whole exome sequencing a novel heterozygous EMILIN1 mutation c.748C>T [p.R250C] located in the coiled coil forming region of the protein, in four affected members of an autosomal dominant family presenting a distal motor neuropathy phenotype. In affected patient a sensory nerve biopsy showed slight and unspecific changes in the number and morphology of myelinated fibers. Immunofluorescence study of a motor nerve within a muscle biopsy documented the presence of EMILIN-1 in nerve structures. Skin section and skin derived fibroblasts displayed a reduced extracellular deposition of EMILIN-1 protein with a disorganized network of poorly ramified fibers in comparison with controls. Downregulation of emilin1a in zebrafish displayed developmental delay, locomotion defects, and abnormal axonal arborization from spinal cord motor neurons. The phenotype was complemented by wild-type zebrafish emilin1a, and partially the human wild-type EMILIN1 cRNA, but not by the cRNA harboring the novel c.748C>T [p.R250C]. These data suggest a role of EMILIN-1 in the pathogenesis of diseases affecting the peripheral nervous system.


Asunto(s)
Fibroblastos/patología , Glicoproteínas de Membrana/genética , Mutación/genética , Piel/patología , Adolescente , Animales , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Adulto Joven , Pez Cebra
11.
Am J Pathol ; 189(2): 354-369, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30448410

RESUMEN

In muscular dystrophies, muscle membrane fragility results in a tissue-specific increase of danger-associated molecular pattern molecules (DAMPs) and infiltration of inflammatory cells. The DAMP extracellular ATP (eATP) released by dying myofibers steadily activates muscle and immune purinergic receptors exerting dual negative effects: a direct damage linked to altered intracellular calcium homeostasis in muscle cells and an indirect toxicity through the triggering of the immune response and inhibition of regulatory T cells. Accordingly, pharmacologic and genetic inhibition of eATP signaling improves the phenotype in models of chronic inflammatory diseases. In α-sarcoglycanopathy, eATP effects may be further amplified because α-sarcoglycan extracellular domain binds eATP and displays an ecto-ATPase activity, thus controlling eATP concentration at the cell surface and attenuating the magnitude and/or the duration of eATP-induced signals. Herein, we show that in vivo blockade of the eATP/P2X purinergic pathway by a broad-spectrum P2X receptor-antagonist delayed the progression of the dystrophic phenotype in α-sarcoglycan-null mice. eATP blockade dampened the muscular inflammatory response and enhanced the recruitment of forkhead box protein P3-positive immunosuppressive regulatory CD4+ T cells. The improvement of the inflammatory features was associated with increased strength, reduced necrosis, and limited expression of profibrotic factors, suggesting that pharmacologic purinergic antagonism, altering the innate and adaptive immune component in muscle infiltrates, might provide a therapeutic approach to slow disease progression in α-sarcoglycanopathy.


Asunto(s)
Adenosina Trifosfato/inmunología , Distrofia Muscular Animal , Miofibrillas , Sarcoglicanos/deficiencia , Linfocitos T Reguladores , Adenosina Trifosfato/genética , Animales , Calcio/inmunología , Enfermedad Crónica , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Noqueados , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/inmunología , Distrofia Muscular Animal/patología , Miofibrillas/inmunología , Miofibrillas/patología , Receptores Purinérgicos P2X/genética , Receptores Purinérgicos P2X/inmunología , Sarcoglicanos/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
12.
Int J Mol Sci ; 21(17)2020 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-32825102

RESUMEN

In muscle ATP is primarily known for its function as an energy source and as a mediator of the "excitation-transcription" process, which guarantees muscle plasticity in response to environmental stimuli. When quickly released in massive concentrations in the extracellular space as in presence of muscle membrane damage, ATP acts as a damage-associated molecular pattern molecule (DAMP). In experimental murine models of muscular dystrophies characterized by membrane instability, blockade of eATP/P2X7 receptor (R) purinergic signaling delayed the progression of the dystrophic phenotype dampening the local inflammatory response and inducing Foxp3+ T Regulatory lymphocytes. These discoveries highlighted the relevance of ATP as a harbinger of immune-tissue damage in muscular genetic diseases. Given the interactions between the immune system and muscle regeneration, the comprehension of ATP/purinerigic pathway articulated organization in muscle cells has become of extreme interest. This review explores ATP release, metabolism, feedback control and cross-talk with members of muscle inflammasome in the context of muscular dystrophies.


Asunto(s)
Adenosina Trifosfato/metabolismo , Inflamasomas/metabolismo , Distrofias Musculares/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Animales , Humanos , Músculo Esquelético/metabolismo , Transducción de Señal
13.
Neurogenetics ; 20(3): 165-172, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31267352

RESUMEN

TSFM is a nuclear gene encoding the elongation factor Ts (EFTs), an essential component of mitochondrial translational machinery. Impaired mitochondrial translation is responsible for neurodegenerative disorders characterized by multiple respiratory chain complex defects, multisystemic involvement, and neuroradiological features of Leigh-like syndrome. With the use of a next-generation sequencing (NGS)-based multigene panel for mitochondrial disorders, we identified the novel TSFM homozygous variant c.547G>A (p.Gly183Ser) in a 5-year-old boy with infantile early onset encephalocardiomyopathy, sensorineural hearing loss, and peculiar partially reversible neuroimaging features. Our findings expand the phenotypic spectrum of TSFM-related encephalopathy, offering new insights into the natural history of brain involvement and suggesting that TSFM should be investigated in pediatric mitochondrial disorders with distinctive neurologic and cardiac involvement.


Asunto(s)
Encefalopatías/genética , Cardiomiopatías/genética , Variación Genética , Pérdida Auditiva Sensorineural/genética , Proteínas Mitocondriales/genética , Factores de Elongación de Péptidos/genética , Biopsia , Encéfalo/diagnóstico por imagen , Encefalopatías/complicaciones , Cardiomiopatías/complicaciones , Preescolar , Discapacidades del Desarrollo/genética , Transporte de Electrón , Pérdida Auditiva Sensorineural/complicaciones , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Imagen por Resonancia Magnética , Masculino , Enfermedades Mitocondriales/genética , Hipotonía Muscular/genética , Neuroimagen , Análisis de Secuencia por Matrices de Oligonucleótidos , Biosíntesis de Proteínas
15.
Am J Med Genet A ; 179(2): 317-321, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30561154

RESUMEN

Kyphoscoliotic Ehlers-Danlos syndrome associated with FKBP14 (FKBP14-kEDS) is an ultrarare autosomal recessive disorder reported in less than 30 individuals so far. In its original description, emphasis was put on the mild muscle involvement. Further reports confirm that FKBP14-kEDS is distinguishable from primary muscle disorders by the lack of progressive muscle disease. We report a 15-year-old girl with FKBP14-kEDS as a result of the recurrent c.362dupC variant, who also showed severe involvement of the lower limb muscles. She never attained autonomous walking and presented significant lower limb weakness. Lower limb magnetic resonance imaging showed a pattern of multiple muscle involvement. Further musculoskeletal assessment revealed significant bone mass density reduction of the spine, unilateral congenital hip dysplasia, and occipitoatlantoaxial instability. This patient points out the existence of a wider phenotypic spectrum of FKBP14-kEDS to include early onset muscle disease.


Asunto(s)
Síndrome de Ehlers-Danlos/genética , Isomerasa de Peptidilprolil/genética , Adolescente , Síndrome de Ehlers-Danlos/diagnóstico por imagen , Síndrome de Ehlers-Danlos/fisiopatología , Femenino , Homocigoto , Humanos , Imagen por Resonancia Magnética , Mutación/genética , Fenotipo
16.
Epilepsia ; 60(5): e31-e36, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30719712

RESUMEN

Juvenile myoclonic epilepsy (JME) is a common syndrome of genetic generalized epilepsies (GGEs). Linkage and association studies suggest that the gene encoding the bromodomain-containing protein 2 (BRD2) may increase risk of JME. The present methylation and association study followed up a recent report highlighting that the BRD2 promoter CpG island (CpG76) is differentially hypermethylated in lymphoblastoid cells from Caucasian patients with JME compared to patients with other GGE subtypes and unaffected relatives. In contrast, we found a uniform low average percentage of methylation (<4.5%) for 13 CpG76-CpGs in whole blood cells from 782 unrelated European Caucasians, including 116 JME patients, 196 patients with genetic absence epilepsies, and 470 control subjects. We also failed to confirm an allelic association of the BRD2 promoter single nucleotide polymorphism (SNP) rs3918149 with JME (Armitage trend test, P = 0.98), and we did not detect a substantial impact of SNP rs3918149 on CpG76 methylation in either 116 JME patients (methylation quantitative trait loci [meQTL], P = 0.29) or 470 German control subjects (meQTL, P = 0.55). Our results do not support the previous observation that a high DNA methylation level of the BRD2 promoter CpG76 island is a prevalent epigenetic motif associated with JME in Caucasians.


Asunto(s)
Islas de CpG/genética , Metilación de ADN , Epilepsia Mioclónica Juvenil/genética , Regiones Promotoras Genéticas/genética , Factores de Transcripción/genética , Epilepsia Tipo Ausencia/epidemiología , Epilepsia Tipo Ausencia/genética , Europa (Continente) , Femenino , Humanos , Leucocitos/química , Masculino , Epilepsia Mioclónica Juvenil/sangre , Epilepsia Mioclónica Juvenil/epidemiología , Polimorfismo de Nucleótido Simple
17.
J Hum Genet ; 63(6): 761-764, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29556034

RESUMEN

We present a 13-year-old patient with persistent increase of serum Creatine Kinase (CK) and myalgia after exertion. Skeletal muscle biopsy showed marked reduction of dystrophin expression leading to genetic analysis of DMD gene by MLPA, which detected a single deletion of exon 78. To the best of our knowledge, DMD exon 78 deletion has never been described in literature and, according to prediction, it should lead to loss of reading frame in the dystrophin gene. To further assess the actual effect of exon 78 deletion, we analysed cDNA from muscle mRNA. This analysis confirmed the absence of 32 bp of exon 78. Exclusion of exon 78 changes the open reading frame of exon 79 and generate a downstream stop codon, producing a dystrophin protein of 3703 amino acids instead of 3685 amino acids. Albeit loss of reading frame usually leads to protein degradation and severe phenotype, in this case, we demonstrated that deletion of DMD exon 78 can be associated with a functional protein able to bind DGC complex and a very mild phenotype. This study adds a novel deletion in DMD gene in human and helps to define the compliance between maintaining/disrupting the reading frame and clinical form of the disease.


Asunto(s)
Creatina Quinasa/sangre , Distrofina/genética , Exones , Eliminación de Gen , Distrofia Muscular de Duchenne/diagnóstico , Adolescente , Biopsia , Codón de Terminación , ADN Complementario/genética , Humanos , Masculino , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , Mialgia/fisiopatología , Sistemas de Lectura Abierta , Fenotipo , ARN Mensajero/genética
18.
Muscle Nerve ; 55(1): 55-68, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27184587

RESUMEN

INTRODUCTION: Limb girdle muscular dystrophies (LGMDs) are characterized by high molecular heterogeneity, clinical overlap, and a paucity of specific biomarkers. Their molecular definition is fundamental for prognostic and therapeutic purposes. METHODS: We created an Italian LGMD registry that included 370 molecularly defined patients. We reviewed detailed retrospective and prospective data and compared each LGMD subtype for differential diagnosis purposes. RESULTS: LGMD types 2A and 2B are the most frequent forms in Italy. The ages at disease onset, clinical progression, and cardiac and respiratory involvement can vary greatly between each LGMD subtype. In a set of extensively studied patients, targeted next-generation sequencing (NGS) identified mutations in 36.5% of cases. CONCLUSION: Detailed clinical characterization combined with muscle tissue analysis is fundamental to guide differential diagnosis and to address molecular tests. NGS is useful for diagnosing forms without specific biomarkers, although, at least in our study cohort, several LGMD disease mechanisms remain to be identified. Muscle Nerve 55: 55-68, 2017.


Asunto(s)
Diagnóstico Diferencial , Distrofia Muscular de Cinturas/diagnóstico , Distrofia Muscular de Cinturas/epidemiología , Adolescente , Adulto , Edad de Inicio , Anciano , Creatina Quinasa/sangre , Femenino , Estudios de Asociación Genética , Humanos , Italia/epidemiología , Masculino , Persona de Mediana Edad , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/complicaciones , Distrofia Muscular de Cinturas/genética , Sistema de Registros , Trastornos Respiratorios/etiología , Estadísticas no Paramétricas , Adulto Joven
19.
Lab Invest ; 96(8): 862-71, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27295345

RESUMEN

Activation of the proteasome pathway is one of the secondary processes of cell damage, which ultimately lead to muscle degeneration and necrosis in Duchenne muscular dystrophy (DMD). In mdx mice, the proteasome inhibitor bortezomib up-regulates the membrane expression of members of the dystrophin complex and reduces the inflammatory reaction. However, chronic inhibition of the 26S proteasome may be toxic, as indicated by the systemic side-effects caused by this drug. Therefore, we sought to determine the components of the ubiquitin-proteasome pathway that are specifically activated in human dystrophin-deficient muscles. The analysis of a cohort of patients with genetically determined DMD or Becker muscular dystrophy (BMD) unveiled a selective up-regulation of the ubiquitin ligase tripartite motif-containing protein 32 (TRIM32). The induction of TRIM32 was due to a transcriptional effect and it correlated with disease severity in BMD patients. In contrast, atrogin1 and muscle RING-finger protein-1 (MuRF-1), which are strongly increased in distinct types of muscular atrophy, were not affected by the DMD dystrophic process. Knock-out models showed that TRIM32 is involved in ubiquitination of muscle cytoskeletal proteins as well as of protein inhibitor of activated STAT protein gamma (Piasγ) and N-myc downstream-regulated gene, two inhibitors of satellite cell proliferation and differentiation. Accordingly, we showed that in DMD/BMD muscle tissue, TRIM32 induction was more pronounced in regenerating myofibers rather than in necrotic muscle cells, thus pointing out a role of this protein in the regulation of human myoblast cell fate. This finding highlights TRIM32 as a possible therapeutic target to favor skeletal muscle regeneration in DMD patients.


Asunto(s)
Distrofia Muscular de Duchenne/metabolismo , Factores de Transcripción/biosíntesis , Proteínas de Motivos Tripartitos/biosíntesis , Ubiquitina-Proteína Ligasas/biosíntesis , Animales , Estudios de Casos y Controles , Humanos , Masculino , Ratones , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Músculo Cuádriceps/metabolismo , Músculo Cuádriceps/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regeneración , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA