Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 235
Filtrar
1.
Dis Model Mech ; 17(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-39034883

RESUMEN

Pontocerebellar hypoplasia type 2a (PCH2a) is an ultra-rare, autosomal recessive pediatric disorder with limited treatment options. Its anatomical hallmark is hypoplasia of the cerebellum and pons accompanied by progressive microcephaly. A homozygous founder variant in TSEN54, which encodes a tRNA splicing endonuclease (TSEN) complex subunit, is causal. The pathological mechanism of PCH2a remains unknown due to the lack of a model system. Therefore, we developed human models of PCH2a using regionalized neural organoids. We generated induced pluripotent stem cell (iPSC) lines from three males with genetically confirmed PCH2a and subsequently differentiated cerebellar and neocortical organoids. Mirroring clinical neuroimaging findings, PCH2a cerebellar organoids were reduced in size compared to controls starting early in differentiation. Neocortical PCH2a organoids demonstrated milder growth deficits. Although PCH2a cerebellar organoids did not upregulate apoptosis, their stem cell zones showed altered proliferation kinetics, with increased proliferation at day 30 and reduced proliferation at day 50 compared to controls. In summary, we generated a human model of PCH2a, providing the foundation for deciphering brain region-specific disease mechanisms. Our first analyses suggest a neurodevelopmental aspect of PCH2a.


Asunto(s)
Encéfalo , Diferenciación Celular , Células Madre Pluripotentes Inducidas , Organoides , Humanos , Organoides/patología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Masculino , Encéfalo/patología , Cerebelo/anomalías , Cerebelo/patología , Atrofias Olivopontocerebelosas/patología , Atrofias Olivopontocerebelosas/genética , Proliferación Celular , Tamaño de los Órganos , Modelos Biológicos , Apoptosis , Enfermedades Cerebelosas
2.
Pediatr Neurol ; 158: 1-10, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38925092

RESUMEN

BACKGROUND: Pontocerebellar hypoplasia type 10 (PCH10) due to CLP1 gene mutations is characterized by structural brain anomalies, progressive microcephaly, severe intellectual and physical disabilities, and spasticity. In this follow-up study, evolution of phenotypic and neurological characteristics of patients with PCH10 is discussed. METHODS: Phenotype, growth parameters, motor functions, developmental tests, spasticity assessments, functional independence assessments, electroencephalography (EEG), and brain magnetic resonance imaging (MRI) of 10 patients with PCH10 were monitored on separate examinations. Alterations were recorded. RESULTS: Patients were followed-up for an average of 2.83 years. The tone of the upper extremities was significantly higher than that of the lower extremities, according to Modified Ashworth Scale (MAS) values. Sixty percent of patients could sit unsupported; 20% achieved supported sitting initially but lost the ability during follow-up. Absence of grabbing or sitting was observed in 20% of patients. During follow-up, one person achieved supported sitting and one person achieved head holding. Only one patient was able to speak a few words. Cerebellar atrophy (two of 10), pons hypoplasia (four of 10), cortical atrophy (seven of 10), enlarged ventricles (10 of 10), thinning of the corpus callosum (10 of 10), hypomyelination (six of 10), and increased white matter signal intensity (six of 10) were the observed MRI findings. CONCLUSIONS: Progressive cerebral and cerebellar atrophy was demonstrated radiologically for the first time in a PCH10 cohort. It is of crucial importance to identify these patients promptly with the help of dysmorphic findings and spasticity being pronounced in the upper extremities. Furthermore, we note that phenotypic and neurological examination findings tend to change slightly over time.


Asunto(s)
Imagen por Resonancia Magnética , Humanos , Masculino , Femenino , Niño , Preescolar , Adolescente , Estudios de Seguimiento , Progresión de la Enfermedad , Lactante , Atrofias Olivopontocerebelosas/patología , Atrofias Olivopontocerebelosas/diagnóstico por imagen , Atrofias Olivopontocerebelosas/fisiopatología , Electroencefalografía , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Fenotipo , Espasticidad Muscular/fisiopatología , Espasticidad Muscular/diagnóstico por imagen , Enfermedades Cerebelosas
3.
Pediatr Neurol ; 131: 30-41, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35468344

RESUMEN

BACKGROUND: Pontocerebellar hypoplasia type 6 (PCH6) is an early-onset encephalopathy with/without mitochondrial respiratory complex defects caused by recessive mutations in mitochondrial arginyl-tRNA synthetase (RARS2). Highly heterogeneous clinical phenotypes and numerous missense variations of uncertain significance make diagnosis difficult. Pathogenesis of PCH6 remains unclear. METHODS: Facial characteristics of patients were assessed. Genetic tests were performed. Structure prediction was based on the template from AlphaFold Protein Structure Database. Expression of mutant RARS2 was tested in HEK293T cells. Patient-derived induced pluripotent stem cells (iPSCs) were detected for human mitochondrial tRNAArg (hmtRNAArg) steady-state level, mitochondrial respiratory complex (MRC) activity, oxygen consumption rate (OCR), extracellular acidification rate (ECAR), mitochondrial membrane potential (MMP), reactive oxygen species (ROS) abundance, and apoptosis level. RESULTS: The three pedigrees were diagnosed as PCH6 caused by compound heterozygous RARS2 variations. Five RARS2 variants were identified: c.3G>C(p.M1?), c.685C>T(p.R229∗), c.1060T>A(p.F354I), c.1210A>G(p.M404V), and c.1369G>A(p.G457R). RARS2 c.3G>C disrupted protein expression. RARS2 c.685C>T created a truncated protein lacking complete catalytic core and anticodon-binding domain. RARS2 c.1060T>A and c.1369G>A were predicted to cause structural abnormality. The hmtRNAArg steady-state abundance in a patient's iPSCs was unaffected. Mitochondrial energy metabolism was normal, including MRC activity, OCR, ECAR, and MMP, while mitochondria-related cellular characteristics, including ROS (P < 0.001) and apoptosis levels (P < 0.001), increased. CONCLUSIONS: This study reports five RARS2 variations among which c.3G>C and c.1060T>A are novel. Summarized facial features of PCH6 patients will facilitate diagnosis. Defective mitochondrial energy metabolism may not be key points, but mitochondria-related abnormal cellular physiology, including apoptosis, may be an underlying pathogenesis.


Asunto(s)
Arginino-ARNt Ligasa , Atrofias Olivopontocerebelosas , Humanos , Arginino-ARNt Ligasa/genética , Células HEK293 , Mutación/genética , Atrofias Olivopontocerebelosas/diagnóstico , Atrofias Olivopontocerebelosas/patología , Especies Reactivas de Oxígeno
4.
J Med Genet ; 59(4): 399-409, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34085948

RESUMEN

BACKGROUND: Pontocerebellar hypoplasias (PCH) comprise a group of genetically heterogeneous disorders characterised by concurrent hypoplasia of the pons and the cerebellum and variable clinical and imaging features. The current classification includes 13 subtypes, with ~20 known causative genes. Attempts have been made to delineate the phenotypic spectrum associated to specific PCH genes, yet clinical and neuroradiological features are not consistent across studies, making it difficult to define gene-specific outcomes. METHODS: We performed deep clinical and imaging phenotyping in 56 probands with a neuroradiological diagnosis of PCH, who underwent NGS-based panel sequencing of PCH genes and MLPA for CASK rearrangements. Next, we conducted a phenotype-based unsupervised hierarchical cluster analysis to investigate associations between genes and specific phenotypic clusters. RESULTS: A genetic diagnosis was obtained in 43 probands (77%). The most common causative gene was CASK, which accounted for nearly half cases (45%) and was mutated in females and occasionally in males. The European founder mutation p.Ala307Ser in TSEN54 and pathogenic variants in EXOSC3 accounted for 18% and 9% of cases, respectively. VLDLR, TOE1 and RARS2 were mutated in single patients. We were able to confirm only few previously reported associations, including jitteriness and clonus with TSEN54 and lower motor neuron signs with EXOSC3. When considering multiple features simultaneously, a clear association with a phenotypic cluster only emerged for EXOSC3. CONCLUSION: CASK represents the major PCH causative gene in Italy. Phenotypic variability associated with the most common genetic causes of PCH is wider than previously thought, with marked overlap between CASK and TSEN54-associated disorders.


Asunto(s)
Enfermedades Cerebelosas , Atrofias Olivopontocerebelosas , Enfermedades Cerebelosas/genética , Cerebelo/diagnóstico por imagen , Cerebelo/patología , Femenino , Humanos , Masculino , Mutación/genética , Proteínas Nucleares/genética , Atrofias Olivopontocerebelosas/diagnóstico , Atrofias Olivopontocerebelosas/genética , Atrofias Olivopontocerebelosas/patología , Fenotipo
5.
Neuropediatrics ; 52(3): 163-169, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33111306

RESUMEN

BACKGROUND: Pontocerebellar hypoplasia (PCH) is a rare group of disorders mainly affecting the cerebellum and pons. Supratentorial structures are variably involved. We assessed brain growth patterns in patients with the most frequent forms of PCH, namely PCH1B (OMIM#614678) and PCH2A (OMIM#277470), since in these types of PCH, pre- and postnatal neurodegeneration is established by neuropathological profiling. To assess the influence of the different pathomechanisms on postnatal growth patterns, we included CASK-associated microcephaly and PCH (MICPCH, OMIM#300749) patients in our analyses, as MICPH mimics PCH on magnetic resonance imaging (MRI) but represents a developmental disorder including abnormal neuronal migration. METHODS: A total of 66 patients were included: 9 patients with PCH1B, 18 patients with PCH2A, 6 patients with MICPCH, and 33 age- and gender-matched hospital-based controls. Segmentation of the vermis and cerebellum was performed manually, as were measurements of the thickness of the head of the caudate nucleus, the width of the anterior horn, and lateral ventricle size. RESULTS: The cerebellum was severely hypoplastic at birth in all patients, and postnatal growth was nearly absent. In patients with PCH1B/2A, we found relative sparing of the vermis compared with the cerebellar hemispheres. In addition, PCH1B and PCH2A cases demonstrated thinning of the head of the caudate nucleus, an associated increase in anterior horn width, and an increase in lateral ventricle size. None of these features were seen in the MICPCH group. CONCLUSIONS: Our findings confirm the progressive nature including caudate nucleus atrophy in PCH1B and PCH2A. In MICPCH, the relative sparing of supratentorial structures confirms its different pathomechanism.


Asunto(s)
Enfermedades Cerebelosas , Atrofias Olivopontocerebelosas , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Enfermedades Cerebelosas/diagnóstico por imagen , Enfermedades Cerebelosas/patología , Cerebelo/patología , Humanos , Recién Nacido , Imagen por Resonancia Magnética , Atrofias Olivopontocerebelosas/diagnóstico por imagen , Atrofias Olivopontocerebelosas/patología
6.
J Hum Genet ; 66(4): 401-407, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33040083

RESUMEN

Pontocerebellar hypoplasia (PCH) is currently classified into 13 subgroups and many gene variants associated with PCH have been identified by next generation sequencing. PCH type 1 is a rare heterogeneous neurodegenerative disorder. The clinical presentation includes early-onset severe developmental delay, progressive motor neuronopathy, and cerebellar and pontine atrophy. Recently two variants in the EXOSC9 gene (MIM: 606180), NM_001034194.1: c.41T>C (p.Leu14Pro) and c.481C>T (p.Arg161*) were identified in four unrelated patients with PCH type 1D (PCH1D) (MIM: 618065). EXOSC9 encodes a component of the exosome complex, which is essential for correct processing and degradation of RNA. We report here two PCH1D families with biallelic EXOSC9 variants: c.239T>G (p.Leu80Arg) and c.484dupA (p.Arg162Lysfs*3) in one family and c.151G>C (p.Gly51Arg) in the other family. Although the patients studied here showed similar clinical features as previously described for PCH1D, relatively greater intellectual development (although still highly restricted) and normal pontine structure were recognized. Our findings expand the clinical consequences of biallelic EXOSC9 variants.


Asunto(s)
Atrofia/patología , Enfermedades Cerebelosas/patología , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Enfermedad de la Neurona Motora/patología , Atrofia Muscular Espinal/patología , Mutación , Atrofias Olivopontocerebelosas/patología , Proteínas de Unión al ARN/genética , Atrofia/complicaciones , Atrofia/genética , Enfermedades Cerebelosas/complicaciones , Enfermedades Cerebelosas/genética , Femenino , Estudios de Asociación Genética , Humanos , Lactante , Masculino , Enfermedad de la Neurona Motora/complicaciones , Enfermedad de la Neurona Motora/genética , Atrofia Muscular Espinal/complicaciones , Atrofia Muscular Espinal/genética , Atrofias Olivopontocerebelosas/complicaciones , Atrofias Olivopontocerebelosas/genética , Linaje
7.
Am J Med Genet A ; 182(11): 2675-2679, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32875707

RESUMEN

The CAMK2B gene encodes the ß-subunit of calcium/calmodulin-dependent protein kinase II (CAMK2), an enzyme that has crucial roles in synaptic plasticity, especially in hippocampal and cerebellar neurons. Heterozygous variants in CAMK2B cause a rare neurodevelopmental disorder, with 40% of the reported cases sharing the same variant: c.416C>T, p.(P139L). This case report describes a 22-year-old patient with this recurrent variant, who presents with severe intellectual disability, absence of language, hypotonia, microcephaly, dysmorphic features, epilepsy, behavioral abnormalities, motor stereotypies, optic atrophy, and progressive cerebellar atrophy. Notably, this patient is the oldest reported so far and allows us to better delineate the clinical phenotype associated with this variant, adding clinical aspects never described before, such as epilepsy, optic atrophy, scoliosis, and neuroradiological changes characterized by progressive cerebellar atrophy.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Epilepsia/patología , Discapacidad Intelectual/patología , Trastornos del Lenguaje/patología , Mutación , Atrofias Olivopontocerebelosas/patología , Adulto , Epilepsia/genética , Femenino , Humanos , Discapacidad Intelectual/genética , Trastornos del Lenguaje/genética , Atrofias Olivopontocerebelosas/genética , Fenotipo , Pronóstico , Adulto Joven
8.
Am J Med Genet A ; 182(10): 2377-2383, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32744787

RESUMEN

Mutations in the MBOAT7 gene have been described in 43 patients, belonging to 18 families, showing nonspecific clinical features (intellectual disability [ID], seizures, microcephaly or macrocephaly, and mild to moderate cerebellar atrophy) that make the clinical diagnosis difficult. Here we report the first Italian patient, a 22.5-year-old female, one of the oldest reported, born to apparently consanguineous parents. She shows severe ID, macrocephaly, seizures, aggressive outbursts, hyperphagia. We also documented progressive atrophy of the cerebellar vermis, that appeared not before the age of 7. The whole-exome sequencing of the trio identified a novel homozygous variant c.1057_1058delGCinsCA (p.Ala353His) in the MBOAT7 gene. The variant is considered to be likely pathogenic, since it is absent from population database and it lies in a highly conserved amino acid residue. This disorder has a neurometabolic pathogenesis, implicating a phospholipid remodeling abnormalities. A brain hydrogen-magnetic resonance spectroscopy (H-MRS) examination in our patient disclosed a peculiar neurometabolic profile in the cerebellar hemispheric region. This new finding could address the clinical suspicion of MBOAT7-related disorder, among the wide range of genetic conditions associated with ID and cerebellar atrophy. Moreover, the documented progression of cerebellar atrophy and the worsening of the disease only after some years open to the possibility of a therapeutic window after birth.


Asunto(s)
Aciltransferasas/genética , Predisposición Genética a la Enfermedad , Discapacidad Intelectual/genética , Proteínas de la Membrana/genética , Atrofias Olivopontocerebelosas/genética , Adolescente , Adulto , Cerebelo/diagnóstico por imagen , Niño , Consanguinidad , Exoma/genética , Femenino , Homocigoto , Humanos , Discapacidad Intelectual/patología , Masculino , Atrofias Olivopontocerebelosas/patología , Linaje , Secuenciación del Exoma , Adulto Joven
9.
Neuropediatrics ; 51(6): 440-444, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32629522

RESUMEN

Pontocerebellar hypoplasia (PCH) is an autosomal recessive neurodevelopmental and neurodegenerative disorder characterized by cerebellar and pontine hypoplasia, progressive microcephaly, and developmental delay. Ten types of PCH have been described; PCH type 2A (PCH2A) due to a mutation in TSEN54 is the most frequent. Seizures have been reported in the large majority of patients. The probability of epilepsy developing increases with age, along with difficulties in differentiating seizures from dyskinetic movements. The aim of the present report was to describe the clinical symptoms and electroencephalogram (EEG) changes over time in three patients of Israeli Arab origin with PCH2A. All three, including two siblings and their first cousin, were homozygous for the TSEN54 p.A304S mutation. The patients demonstrated profound psychomotor retardation, severe spasticity and contractures, choreoathetoid movements, and seizures. The magnetic resonance imaging (MRI) scans and EEGs were reviewed by an experienced neuroradiologist and epileptologist, respectively. The MRI scans revealed a dragonfly-like cerebellar pattern in all patients. Despite the normal early EEG findings, all patients had characteristic features of epilepsy, with tonic seizures starting in the first days to months followed by focal to bilateral tonic-clonic seizures in early childhood which continued to adolescence. In conclusion, patients with PCH2A due to the missense mutation p.A304S in TSEN54 exhibit profound psychomotor delay, movement disorders, and intractable epilepsy. An evolution of EEG abnormalities and seizure semiology occurs over time. Similar to several other genetic epileptic encephalopathies, the normal early EEG tracing does not rule out the later occurrence of epilepsy.


Asunto(s)
Encéfalo/patología , Encéfalo/fisiopatología , Atrofias Olivopontocerebelosas/patología , Atrofias Olivopontocerebelosas/fisiopatología , Adolescente , Niño , Progresión de la Enfermedad , Electroencefalografía , Femenino , Humanos
10.
Parkinsonism Relat Disord ; 73: 94-104, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32005598

RESUMEN

Multiple system atrophy (MSA) is a rare and fatal neurodegenerative disorder characterized by rapidly progressive autonomic and motor dysfunction. Pathologically, MSA is mainly characterized by the abnormal accumulation of misfolded α-synuclein in the cytoplasm of oligodendrocytes, which plays a major role in the pathogenesis of the disease. Striatonigral degeneration and olivopontecerebellar atrophy underlie the motor syndrome, while degeneration of autonomic centers defines the autonomic failure in MSA. At present, there is no treatment that can halt or reverse its progression. However, over the last decade several studies in preclinical models and patients have helped to better understand the pathophysiological events underlying MSA. The etiology of this fatal disorder remains unclear and may be multifactorial, caused by a combination of factors which may serve as targets for novel therapeutic approaches. In this review, we summarize the current knowledge about the etiopathogenesis and neuropathology of MSA, its different preclinical models, and the main disease modifying therapies that have been used so far or that are planned for future clinical trials.


Asunto(s)
Atrofia de Múltiples Sistemas , Atrofias Olivopontocerebelosas , Degeneración Estriatonigral , Animales , Humanos , Atrofia de Múltiples Sistemas/tratamiento farmacológico , Atrofia de Múltiples Sistemas/etiología , Atrofia de Múltiples Sistemas/patología , Atrofias Olivopontocerebelosas/tratamiento farmacológico , Atrofias Olivopontocerebelosas/etiología , Atrofias Olivopontocerebelosas/patología , Degeneración Estriatonigral/tratamiento farmacológico , Degeneración Estriatonigral/etiología , Degeneración Estriatonigral/patología
11.
Eur J Med Genet ; 63(1): 103622, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30690203

RESUMEN

Pontocerebellar Hypoplasia type 1 is a rare heterogeneous neurodegenerative disorder with multiple subtypes linked to dysfunction of the exosome complex. Patients with mutations in exosome subunits exhibit a generally lethal phenotype characterized by cerebellar and pontine hypoplasia in association with spinal motor neuropathy and multiple systemic and neurologic features. Recently, two variants in the novel PCH1 associated protein EXOSC9 p.(Leu14Pro) and p.(Arg161*) have been identified in 4 unrelated patients exhibiting a severe phenotype involving cerebellar hypoplasia, axonal motor neuropathy, hypotonia, feeding difficulties, and respiratory insufficiency (PCH1D). We report clinical and molecular characterization of 2 unrelated patients exhibiting a relatively milder phenotype involving hypotonia, brachycephaly, cerebellar atrophy, psychomotor delay, as well as lactic acidosis and aberrant CNS myelination, resulting from the recurring homozygous missense mutation NM_001034194.1: c.41T>C; p.(Leu14Pro) in the EXOSC9 gene. We review the clinical picture of the EXOSC9-related PCH disorder.


Asunto(s)
Cerebelo/anomalías , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Predisposición Genética a la Enfermedad , Malformaciones del Sistema Nervioso/genética , Atrofias Olivopontocerebelosas/genética , Proteínas de Unión al ARN/genética , Neoplasias del Tronco Encefálico/genética , Neoplasias del Tronco Encefálico/patología , Cerebelo/patología , Niño , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Femenino , Estudios de Asociación Genética , Homocigoto , Humanos , Masculino , Mutación/genética , Malformaciones del Sistema Nervioso/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Atrofias Olivopontocerebelosas/patología , Fenotipo , Nervios Espinales/patología
12.
Muscle Nerve ; 59(1): 137-141, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30025162

RESUMEN

INTRODUCTION: Mutations in the EXOSC3 gene are responsible for type 1 pontocerebellar hypoplasia, an autosomal recessive congenital disorder characterized by cerebellar atrophy, developmental delay, and anterior horn motor neuron degeneration. Muscle biopsies of these patients often show characteristics resembling classic spinal muscle atrophy, but to date, no distinct features have been identified. METHODS: Clinical data and muscle biopsy findings of 3 unrelated patients with EXOSC3 mutations are described. RESULTS: All patients presented as a severe congenital cognitive and neuromuscular phenotype with short survival, harboring the same point mutation (c.92G>C; p.Gly31Ala). Muscle biopsies consistently showed variable degrees of sarcomeric disorganization with myofibrillar remnants, Z-line thickening, and small nemaline bodies. CONCLUSIONS: In this uniform genetic cohort of patients with EXOSC3 mutations, sarcomeric disruption and rod structures were prominent features of muscle biopsies. In the context of neonatal hypotonia, ultrastructural studies might provide early clues for the diagnosis of EXOSC3-related pontocerebellar hypoplasia. Muscle Nerve 59:137-141, 2019.


Asunto(s)
Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Músculo Esquelético/patología , Mutación/genética , Atrofias Olivopontocerebelosas/genética , Atrofias Olivopontocerebelosas/patología , Proteínas de Unión al ARN/genética , Sarcoma/patología , Biopsia , Preescolar , Estudios de Cohortes , Femenino , Humanos , Recién Nacido , Masculino , Músculo Esquelético/ultraestructura , Miopatías Nemalínicas , Sarcoma/ultraestructura
13.
ACS Chem Biol ; 13(10): 3000-3010, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30141626

RESUMEN

Mutations of EXOSC3 have been linked to the rare neurological disorder known as Pontocerebellar Hypoplasia type 1B (PCH1B). EXOSC3 is one of three putative RNA-binding structural cap proteins that guide RNA into the RNA exosome, the cellular machinery that degrades RNA. Using RNAcompete, we identified a G-rich RNA motif binding to EXOSC3. Surface plasmon resonance (SPR) and microscale thermophoresis (MST) indicated an affinity in the low micromolar range of EXOSC3 for long and short G-rich RNA sequences. Although several PCH1B-causing mutations in EXOSC3 did not engage a specific RNA motif as shown by RNAcompete, they exhibited lower binding affinity to G-rich RNA as demonstrated by MST. To test the hypothesis that modification of the RNA-protein interface in EXOSC3 mutants may be phenocopied by small molecules, we performed an in-silico screen of 50 000 small molecules and used enzyme-linked immunosorbant assays (ELISAs) and MST to assess the ability of the molecules to inhibit RNA-binding by EXOSC3. We identified a small molecule, EXOSC3-RNA disrupting (ERD) compound 3 (ERD03), which ( i) bound specifically to EXOSC3 in saturation transfer difference nuclear magnetic resonance (STD-NMR), ( ii) disrupted the EXOSC3-RNA interaction in a concentration-dependent manner, and ( iii) produced a PCH1B-like phenotype with a 50% reduction in the cerebellum and an abnormally curved spine in zebrafish embryos. This compound also induced modification of zebrafish RNA expression levels similar to that observed with a morpholino against EXOSC3. To our knowledge, this is the first example of a small molecule obtained by rational design that models the abnormal developmental effects of a neurodegenerative disease in a whole organism.


Asunto(s)
Modelos Animales de Enfermedad , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Isoquinolinas/farmacología , Isoquinolinas/toxicidad , Atrofias Olivopontocerebelosas/genética , Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Pez Cebra/anomalías , Animales , Atrofia , Cerebelo/patología , Regulación hacia Abajo , Complejo Multienzimático de Ribonucleasas del Exosoma/química , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Técnicas de Silenciamiento del Gen , Humanos , Isoquinolinas/metabolismo , Simulación del Acoplamiento Molecular , Mutación , Atrofias Olivopontocerebelosas/inducido químicamente , Atrofias Olivopontocerebelosas/patología , Fenotipo , Unión Proteica , Dominios Proteicos , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Curvaturas de la Columna Vertebral/inducido químicamente , Transcriptoma/efectos de los fármacos , Regulación hacia Arriba
14.
J Biol Chem ; 293(35): 13604-13615, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-30006346

RESUMEN

Human mitochondrial aminoacyl-tRNA synthetases (mt-aaRSs) are key enzymes in the mitochondrial protein translation system and catalyze the charging of amino acids on their cognate tRNAs. Mutations in their nuclear genes are associated with pathologies having a broad spectrum of clinical phenotypes, but with no clear molecular mechanism(s). For example, mutations in the nuclear genes encoding mt-AspRS and mt-ArgRS are correlated with the moderate neurodegenerative disorder leukoencephalopathy with brainstem and spinal cord involvement and lactate elevation (LBSL) and with the severe neurodevelopmental disorder pontocerebellar hypoplasia type 6 (PCH6), respectively. Previous studies have shown no or only minor impacts of these mutations on the canonical properties of these enzymes, indicating that the role of the mt-aaRSs in protein synthesis is mostly not affected by these mutations, but their effects on the mitochondrial localizations of aaRSs remain unclear. Here, we demonstrate that three human aaRSs, mt-AspRS, mt-ArgRS, and LysRS, each have a specific sub-mitochondrial distribution, with mt-ArgRS being exclusively localized in the membrane, LysRS exclusively in the soluble fraction, and mt-AspRS being present in both. Chemical treatments revealed that mt-AspRs is anchored in the mitochondrial membrane through electrostatic interactions, whereas mt-ArgRS uses hydrophobic interactions. We also report that novel mutations in mt-AspRS and mt-ArgRS genes from individuals with LBSL and PCH6, respectively, had no significant impact on the mitochondrial localizations of mt-AspRS and mt-ArgRS. The variable sub-mitochondrial locations for these three mt-aaRSs strongly suggest the existence of additional enzyme properties, requiring further investigation to unravel the mechanisms underlying the two neurodegenerative disorders.


Asunto(s)
Arginino-ARNt Ligasa/análisis , Aspartato-ARNt Ligasa/análisis , Lisina-ARNt Ligasa/análisis , Mitocondrias/química , Arginino-ARNt Ligasa/genética , Aspartato-ARNt Ligasa/genética , Femenino , Células HEK293 , Humanos , Leucoencefalopatías/genética , Leucoencefalopatías/patología , Lisina-ARNt Ligasa/genética , Mitocondrias/genética , Mitocondrias/patología , Mutación , Atrofias Olivopontocerebelosas/genética , Atrofias Olivopontocerebelosas/patología
15.
J Neuropathol Exp Neurol ; 77(7): 598-607, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29850876

RESUMEN

Multiple system atrophy (MSA) is an adult-onset neurodegenerative disease characterized by aggregation of α-synuclein in oligodendrocytes to form glial cytoplasmic inclusions. According to the distribution of neurodegeneration, MSA is subtyped as striatonigral degeneration (SND), olivopontocerebellar atrophy (OPCA), or as combination of these 2 (mixed MSA). In the current study, we aimed to investigate regional microglial populations and gene expression in the 3 different MSA subtypes. Microscopy with microglial marker Iba-1 combined with either proinflammatory marker CD68 or anti-inflammatory marker Arginase-1 was analyzed in control, SND, and OPCA cases (n = 5) using paraffin embedded sections. Western immunoblotting and cytokine array were used to determine protein expression in MSA and control brain regions. Gene expression was investigated using the NanoString nCounter Human Inflammation panel v2 mRNA Expression Assay. Analysis of neuropathological subtypes of MSA demonstrated a significant increase in microglia in the substantia nigra of OPCA cases. There was no difference in the microglial activation state in any region. Cytokine expression in MSA was comparable with controls. Decreased expression of CX3CL1 precursor protein and significantly greater CX3CR1 protein was found in MSA. NanoString analysis revealed the >2-fold greater expression of ARG1, MASP1, NOX4, PTGDR2, and C6 in MSA.


Asunto(s)
Encéfalo/patología , Inflamación/genética , Inflamación/patología , Atrofia de Múltiples Sistemas/genética , Atrofia de Múltiples Sistemas/patología , Antígenos CD/biosíntesis , Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/biosíntesis , Antígenos de Diferenciación Mielomonocítica/genética , Arginasa/biosíntesis , Arginasa/genética , Proteínas de Unión al Calcio , Quimiocinas/análisis , Quimiocinas/biosíntesis , Cuerpo Estriado/patología , Citocinas/análisis , Citocinas/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/genética , Humanos , Inmunohistoquímica , Proteínas de Microfilamentos , Microglía/patología , Atrofias Olivopontocerebelosas/patología , Sustancia Negra/patología
16.
Eur J Paediatr Neurol ; 22(4): 674-681, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29656927

RESUMEN

Pontocerebellar hypoplasia type 1 (PCH1) is a major cause of non-5q spinal muscular atrophy (SMA). We screened 128 SMN1-negative SMA patients from Bulgaria for a frequent mutation -p.G31A in EXOSC3, and performed a literature review of all genetically verified PCH1 cases. Homozygous p.G31A/EXOSC3 mutation was identified in 14 Roma patients, representing three fourths of all our SMN1-negative Roma SMA cases. The phenotype of the p.G31A/EXOSC3 homozygotes was compared to the clinical presentation of all reported to date genetically verified PCH1 cases. Signs of antenatal onset of disease present at birth were common in all PCH1 sub-types except in the homozygous p.D132A/EXOSC3 patients. The PCH1sub-types with early death (between ages 1 day and 17 months), seen in patients with p.G31A/EXOSC3 or SLC25A46 mutations have a SMA type 1-like clinical presentation but with global developmental delay, visual and hearing impairment, with or without microcephaly, nystagmus and optic atrophy. Mutations with milder presentation (homozygous p.D132A/EXOSC3 or VRK1) may display additionally signs of upper motor neuron impairment, dystonia or ataxia and die at age between 5 and 18 years. Other EXOSC3 mutations and EXOSC8 cases are intermediate - SMA type 1-like presentation, spasticity (mostly in EXOSC8) and death between 3 months and 5 years. There is no correlation between neurological onset and duration of life. We add marble-like skin and congenital laryngeal stridor as features of PCH1. We show that imaging signs of cerebellar and pontine hypoplasia may be missing early in infancy. EMG signs of anterior horn neuronopathy may be missing in PCH1 patients with SLC25A46 mutations. Thus, there is considerable phenotypic variability in PCH1, with some cases being more SMA-like, than PCH-like. Detailed clinical evaluation and ethnicity background may guide genetic testing and subsequent genetic counseling.


Asunto(s)
Atrofias Olivopontocerebelosas/diagnóstico , Atrofias Olivopontocerebelosas/genética , Atrofias Musculares Espinales de la Infancia/genética , Adolescente , Bulgaria , Niño , Preescolar , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Femenino , Estudios de Asociación Genética , Homocigoto , Humanos , Masculino , Mutación , Atrofias Olivopontocerebelosas/patología , Fenotipo , Proteínas de Unión al ARN/genética , Romaní/genética
17.
Mitochondrion ; 37: 46-54, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28687512

RESUMEN

Recessive mutations in EXOSC3, encoding a subunit of the human RNA exosome complex, cause pontocerebellar hypoplasia type 1b (PCH1B). We report a boy with severe muscular hypotonia, psychomotor retardation, progressive microcephaly, and cerebellar atrophy. Biochemical abnormalities comprised mitochondrial complex I and pyruvate dehydrogenase complex (PDHc) deficiency. Whole exome sequencing uncovered a known EXOSC3 mutation p.(D132A) as the underlying cause. In patient fibroblasts, a large portion of the EXOSC3 protein was trapped in the cytosol. MtDNA copy numbers in muscle were reduced to 35%, but mutations in the mtDNA and in nuclear mitochondrial genes were ruled out. RNA-Seq of patient muscle showed highly increased mRNA copy numbers, especially for genes encoding structural subunits of OXPHOS complexes I, III, and IV, possibly due to reduced degradation by a dysfunctional exosome complex. This is the first case of mitochondrial dysfunction associated with an EXOSC3 mutation, which expands the phenotypic spectrum of PCH1B. We discuss the links between exosome and mitochondrial dysfunction.


Asunto(s)
Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Mutación , Atrofias Olivopontocerebelosas/genética , Atrofias Olivopontocerebelosas/patología , Proteínas de Unión al ARN/genética , Complejo I de Transporte de Electrón/deficiencia , Humanos , Lactante , Masculino , Enfermedad por Deficiencia del Complejo Piruvato Deshidrogenasa
18.
RNA ; 23(4): 466-472, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28053271

RESUMEN

The RNA exosome is a conserved multiprotein complex that achieves a large number of processive and degradative functions in eukaryotic cells. Recently, mutations have been mapped to the gene encoding one of the subunits of the exosome, EXOSC3 (yeast Rrp40p), which results in pontocerebellar hypoplasia with motor neuron degeneration in human patients. However, the molecular impact of these mutations in the pathology of these diseases is not well understood. To investigate the molecular consequences of mutations in EXOSC3 that lead to neurological diseases, we analyzed the effect of three of the mutations that affect conserved residues of EXOSC3/Rrp40p (G31A, G191C, and W238R; G8A, G148C, and W195R, respectively, in human and yeast) in S. cerevisiae We show that the severity of the phenotypes of these mutations in yeast correlate with that of the disease in human patients, with the W195R mutant showing the strongest growth and RNA processing phenotypes. Furthermore, we show that these mutations affect more severely pre-ribosomal RNA processing functions of the exosome rather than other nuclear processing or surveillance functions. These results suggest that delayed or defective pre-rRNA processing might be the primary defect responsible for the pathologies detected in patients with mutations affecting EXOSC3 function in residues conserved throughout eukaryotes.


Asunto(s)
Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Mutación , Precursores del ARN/genética , Procesamiento Postranscripcional del ARN , ARN de Hongos/genética , ARN Ribosómico/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión , Secuencia Conservada , Complejo Multienzimático de Ribonucleasas del Exosoma/metabolismo , Regulación Fúngica de la Expresión Génica , Humanos , Atrofias Olivopontocerebelosas/genética , Atrofias Olivopontocerebelosas/metabolismo , Atrofias Olivopontocerebelosas/patología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Precursores del ARN/metabolismo , ARN de Hongos/metabolismo , ARN Ribosómico/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido
19.
Orphanet J Rare Dis ; 11(1): 140, 2016 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-27769281

RESUMEN

BACKGROUND: Pontocerebellar hypoplasia type 6 (PCH6) is a mitochondrial disease caused by mutations in the RARS2 gene. RARS2 encodes mitochondrial arginyl transfer RNA synthetase, an enzyme involved in mitochondrial protein translation. A total of 27 patients from 14 families have been reported so far. Characteristic clinical features comprise neonatal lactic acidosis, severe encephalopathy, intractable seizures, feeding problems and profound developmental delay. Most patients show typical neuroradiologic abnormalities including cerebellar hypoplasia and progressive pontocerebellar atrophy. METHODS: We describe the clinical, biochemical and molecular features of 2 siblings with a novel homozygous mutation in RARS2. Both patients presented neonatally with lactic acidosis. While the older sibling had severe neurological symptoms with microcephaly, seizures and developmental delay, the younger patient was still neurologically asymptomatic at the age of 2 months. RESULTS: MRI studies in both children lacked pontocerebellar involvement. The expression of the OXPHOS complex proteins was decreased in both patients, whereas oxygen consumption was increased. CONCLUSIONS: Characteristic neuroradiological abnormalities of PCH6 such as vermis and cerebellar hypoplasia and progressive pontocerebellar atrophy may be missing in patients with RARS2 mutations. RARS2 testing should therefore also be performed in patients without pontocerebellar hypoplasia but otherwise typical clinical symptoms.


Asunto(s)
Arginino-ARNt Ligasa/genética , Enfermedades Mitocondriales/genética , Mutación/genética , Atrofias Olivopontocerebelosas/genética , Hermanos , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Imagen por Resonancia Magnética , Masculino , Enfermedades Mitocondriales/patología , Atrofias Olivopontocerebelosas/patología
20.
Orphanet J Rare Dis ; 11(1): 100, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27430971

RESUMEN

BACKGROUND: Pontocerebellar hypoplasia type 2 (PCH2) is caused by a defect in the TSEN54-gene and leads to severe and early disruption of brain development, especially of cerebellum and pons. The aim of this work was to quantify the infra- and supratentorial brain growth during postnatal brain development in children with PCH2. METHODS: MRI data of 24 children with PCH2 (age 0.02-17 years., 13 females) were analysed volumetrically and compared to images of 24 typically developing age- and gender-matched children. All children with PCH2 had the homozygous p.A307S mutation in the TSEN54-gene. In 5 patients follow-up MRI investigations were available. Images of the children with PCH2 were available either on film (n = 12) or in digital format (n = 21). Images on film were digitalized. Brain structures were manually masked and further adjusted semi-automatically using intensity thresholding to exclude CSF. Volumes of cerebellum, brain stem, and pons were measured, as well as supratentorial brain and frontal lobe volume. For validation of the method part of the digital images were processed as images on film. In addition, intra- and inter-rater variabilities were tested. RESULTS: Children with PCH2 showed reduced volumes of all measured brain structures compared to healthy controls. Severely hypoplastic cerebellum, pons and brain stem only slightly increased in size postnatally. Supratentorial brain volume also showed reduced growth compared to the healthy controls. Differences between patients and controls could already be seen at birth but became more significant during childhood. Validation of the method showed high precision and reproducibility. CONCLUSIONS: In a genetically very homogenous group of children with PCH2 severely hypoplastic infratentorial structures, the hallmark of the disease, showed only slight increase in volume postnatally. Supratentorial brain structures, which are considered normal at birth, also showed smaller volumes neonatally and a lower growth rate compared to controls, leading to severe microcephaly. Volume loss, however, could not be observed during the first years of life. This argues for a severe disruption of the cerebellar-cerebral networks during pre- and postnatal development caused by a primary cerebellar dysfunction, rather than postnatal neurodegeneration. The developmental progress in these children, although little, further supports this.


Asunto(s)
Encéfalo/patología , Atrofias Olivopontocerebelosas/patología , Adolescente , Tronco Encefálico/patología , Estudios de Casos y Controles , Cerebelo/patología , Niño , Preescolar , Femenino , Lóbulo Frontal/patología , Humanos , Lactante , Recién Nacido , Discapacidad Intelectual/patología , Imagen por Resonancia Magnética , Masculino , Microcefalia/patología , Puente/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...