Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Genes (Basel) ; 15(4)2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38674434

RESUMEN

Oxidative phosphorylation involves a complex multi-enzymatic mitochondrial machinery critical for proper functioning of the cell, and defects herein cause a wide range of diseases called "primary mitochondrial disorders" (PMDs). Mutations in about 400 nuclear and 37 mitochondrial genes have been documented to cause PMDs, which have an estimated birth prevalence of 1:5000. Here, we describe a 4-year-old female presenting from early childhood with psychomotor delay and white matter signal changes affecting several brain regions, including the brainstem, in addition to lactic and phytanic acidosis, compatible with Leigh syndrome, a genetically heterogeneous subgroup of PMDs. Whole genome sequencing of the family trio identified a homozygous 12.9 Kb deletion, entirely overlapping the NDUFA4 gene. Sanger sequencing of the breakpoints revealed that the genomic rearrangement was likely triggered by Alu elements flanking the gene. NDUFA4 encodes for a subunit of the respiratory chain Complex IV, whose activity was significantly reduced in the patient's fibroblasts. In one family, dysfunction of NDUFA4 was previously documented as causing mitochondrial Complex IV deficiency nuclear type 21 (MC4DN21, OMIM 619065), a relatively mild form of Leigh syndrome. Our finding confirms the loss of NDUFA4 function as an ultra-rare cause of Complex IV defect, clinically presenting as Leigh syndrome.


Asunto(s)
Complejo I de Transporte de Electrón , Enfermedad de Leigh , Humanos , Enfermedad de Leigh/genética , Enfermedad de Leigh/patología , Femenino , Preescolar , Complejo IV de Transporte de Electrones/genética , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Linaje , Eliminación de Secuencia
2.
Genes (Basel) ; 14(11)2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-38002928

RESUMEN

Strømme syndrome is an ultra-rare primary ciliopathy with clinical variability. The syndrome is caused by bi-allelic variants in CENPF, a protein with key roles in both chromosomal segregation and ciliogenesis. We report three unrelated patients with Strømme syndrome and, using high-throughput sequencing approaches, we identified novel pathogenic variants in CENPF, including one structural variant, giving a genetic diagnosis to the patients. Patient 1 was a premature baby who died at 26 days with congenital malformations affecting many organs including the brain, eyes, and intestine. She was homozygous for a donor splice variant in CENPF, NM_016343.3:c.1068+1G>A, causing skipping of exon 7, resulting in a frameshift. Patient 2 was a female with intestinal atresia, microcephaly, and a Peters anomaly. She had normal developmental milestones at the age of 7 years. She is compound heterozygous for CENPF NM_016343.3:c.5920dup and c.8991del, both frameshift. Patient 3 was a male with anomalies of the brain, eye, intestine, and kidneys. He was compound heterozygous for CENPF p.(Glu298Ter), and a 5323 bp deletion covering exon 1. CENPF exon 1 is flanked by repetitive sequences that may represent a site of a recurrent structural variation, which should be a focus in patients with Strømme syndrome of unknown etiology.


Asunto(s)
Atresia Intestinal , Microcefalia , Niño , Femenino , Humanos , Lactante , Masculino , Segmento Anterior del Ojo , Atresia Intestinal/genética , Microcefalia/genética , Mutación
3.
Brain ; 146(8): 3513-3527, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36917474

RESUMEN

RNA polymerase I transcribes ribosomal DNA to produce precursor 47S rRNA. Post-transcriptional processing of this rRNA generates mature 28S, 18S and 5.8S rRNAs, which form the ribosomes, together with 5S rRNA, assembly factors and ribosomal proteins. We previously reported a homozygous variant in the catalytic subunit of RNA polymerase I, POLR1A, in two brothers with leukodystrophy and progressive course. However, the disease mechanism remained unknown. In this report, we describe another missense variant POLR1A NM_015425.3:c.1925C>A; p.(Thr642Asn) in homozygosity in two unrelated patients. Patient 1 was a 16-year-old male and Patient 2 was a 2-year-old female. Both patients manifested neurological deficits, with brain MRIs showing hypomyelinating leukodystrophy and cerebellar atrophy; and in Patient 1 additionally with hypointensity of globi pallidi and small volume of the basal ganglia. Patient 1 had progressive disease course, leading to death at the age of 16.5 years. Extensive in vitro experiments in fibroblasts from Patient 1 documented that the mutated POLR1A led to aberrant rRNA processing and degradation, and abnormal nucleolar homeostasis. Proteomics data analyses and further in vitro experiments documented abnormal protein homeostasis, and endoplasmic reticulum stress responses. We confirm that POLR1A biallelic variants cause neurodegenerative disease, expand the knowledge of the clinical phenotype of the disorder, and provide evidence for possible pathological mechanisms leading to POLR1A-related leukodystrophy.


Asunto(s)
Enfermedades Neurodegenerativas , ARN Polimerasa I , Masculino , Femenino , Humanos , ARN Polimerasa I/genética , ARN Polimerasa I/metabolismo , Enfermedades Neurodegenerativas/genética , Proteostasis , ARN Ribosómico/metabolismo , Ribosomas , Procesamiento Postranscripcional del ARN
4.
Sci Signal ; 16(771): eadd0509, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36749824

RESUMEN

Stormorken syndrome is a multiorgan hereditary disease caused by dysfunction of the endoplasmic reticulum (ER) Ca2+ sensor protein STIM1, which forms the Ca2+ release-activated Ca2+ (CRAC) channel together with the plasma membrane channel Orai1. ER Ca2+ store depletion activates STIM1 by releasing the intramolecular "clamp" formed between the coiled coil 1 (CC1) and CC3 domains of the protein, enabling the C terminus to extend and interact with Orai1. The most frequently occurring mutation in patients with Stormorken syndrome is R304W, which destabilizes and extends the STIM1 C terminus independently of ER Ca2+ store depletion, causing constitutive binding to Orai1 and CRAC channel activation. We found that in cis deletion of one amino acid residue, Glu296 (which we called E296del) reversed the pathological effects of R304W. Homozygous Stim1 E296del+R304W mice were viable and phenotypically indistinguishable from wild-type mice. NMR spectroscopy, molecular dynamics simulations, and cellular experiments revealed that although the R304W mutation prevented CC1 from interacting with CC3, the additional deletion of Glu296 opposed this effect by enabling CC1-CC3 binding and restoring the CC domain interactions within STIM1 that are critical for proper CRAC channel function. Our results provide insight into the activation mechanism of STIM1 by clarifying the molecular basis of mutation-elicited protein dysfunction and pathophysiology.


Asunto(s)
Canales de Calcio Activados por la Liberación de Calcio , Proteínas de la Membrana , Ratones , Animales , Proteínas de la Membrana/metabolismo , Canales de Calcio/metabolismo , Aminoácidos/metabolismo , Mutación , Retículo Endoplásmico/metabolismo , Molécula de Interacción Estromal 1/genética , Canales de Calcio Activados por la Liberación de Calcio/genética , Proteína ORAI1/metabolismo , Calcio/metabolismo
5.
Genet Med ; 25(1): 90-102, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36318270

RESUMEN

PURPOSE: Brain monoamine vesicular transport disease is an infantile-onset movement disorder that mimics cerebral palsy. In 2013, the homozygous SLC18A2 variant, p.Pro387Leu, was first reported as a cause of this rare disorder, and dopamine agonists were efficient for treating affected individuals from a single large family. To date, only 6 variants have been reported. In this study, we evaluated genotype-phenotype correlations in individuals with biallelic SLC18A2 variants. METHODS: A total of 42 affected individuals with homozygous SLC18A2 variant alleles were identified. We evaluated genotype-phenotype correlations and the missense variants in the affected individuals based on the structural modeling of rat VMAT2 encoded by Slc18a2, with cytoplasm- and lumen-facing conformations. A Caenorhabditis elegans model was created for functional studies. RESULTS: A total of 19 homozygous SLC18A2 variants, including 3 recurrent variants, were identified using exome sequencing. The affected individuals typically showed global developmental delay, hypotonia, dystonia, oculogyric crisis, and autonomic nervous system involvement (temperature dysregulation/sweating, hypersalivation, and gastrointestinal dysmotility). Among the 58 affected individuals described to date, 16 (28%) died before the age of 13 years. Of the 17 patients with p.Pro237His, 9 died, whereas all 14 patients with p.Pro387Leu survived. Although a dopamine agonist mildly improved the disease symptoms in 18 of 21 patients (86%), some affected individuals with p.Ile43Phe and p.Pro387Leu showed milder phenotypes and presented prolonged survival even without treatment. The C. elegans model showed behavioral abnormalities. CONCLUSION: These data expand the phenotypic and genotypic spectra of SLC18A2-related disorders.


Asunto(s)
Encefalopatías , Distonía , Trastornos del Movimiento , Humanos , Animales , Ratas , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/genética , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Trastornos del Movimiento/genética , Aminas , Encéfalo/metabolismo
6.
Brain ; 145(7): 2602-2616, 2022 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-35104841

RESUMEN

Bi-allelic pathogenic variants in ZBTB11 have been associated with intellectual developmental disorder, autosomal recessive 69 (MRT69; OMIM 618383). We report five patients from three families with novel, bi-allelic variants in ZBTB11. We have expanded the clinical phenotype of MRT69, documenting varied severity of atrophy affecting different brain regions and described combined malonic and methylmalonic aciduria as a biochemical manifestation. As ZBTB11 encodes for a transcriptional regulator, we performeded chromatin immunoprecipitation-sequencing targeting ZBTB11 in fibroblasts from patients and controls. Chromatin immunoprecipitation-sequencing revealed binding of wild-type ZBTB11 to promoters in 238 genes, among which genes encoding proteins involved in mitochondrial functions and RNA processing are over-represented. Mutated ZBTB11 showed reduced binding to 61 of the targeted genes, indicating that the variants act as loss of function. Most of these genes are related to mitochondrial functions. Transcriptome analysis of the patient fibroblasts revealed dysregulation of mitochondrial functions. In addition, we uncovered that reduced binding of the mutated ZBTB11 to ACSF3 leads to decreased ACSF3 transcript level, explaining combined malonic and methylmalonic aciduria. Collectively, these results expand the clinical spectrum of ZBTB11-related neurological disease and give insight into the pathophysiology in which the dysfunctional ZBTB11 affect mitochondrial functions and RNA processing contributing to the neurological and biochemical phenotypes.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos , Errores Innatos del Metabolismo , Malformaciones del Sistema Nervioso , Errores Innatos del Metabolismo de los Aminoácidos/genética , Encéfalo , Humanos , Errores Innatos del Metabolismo/genética
7.
Genet Med ; 24(4): 905-914, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35027293

RESUMEN

PURPOSE: Gabriele-de Vries syndrome (GADEVS) is a rare genetic disorder characterized by developmental delay and/or intellectual disability, hypotonia, feeding difficulties, and distinct facial features. To refine the phenotype and to better understand the molecular basis of the syndrome, we analyzed clinical data and performed genome-wide DNA methylation analysis of a series of individuals carrying a YY1 variant. METHODS: Clinical data were collected for 13 individuals not yet reported through an international call for collaboration. DNA was collected for 11 of these individuals and 2 previously reported individuals in an attempt to delineate a specific DNA methylation signature in GADEVS. RESULTS: Phenotype in most individuals overlapped with the previously described features. We described 1 individual with atypical phenotype, heterozygous for a missense variant in a domain usually not involved in individuals with YY1 pathogenic missense variations. We also described a specific peripheral blood DNA methylation profile associated with YY1 variants. CONCLUSION: We reported a distinct DNA methylation episignature in GADEVS. We expanded the clinical profile of GADEVS to include thin/sparse hair and cryptorchidism. We also highlighted the utility of DNA methylation episignature analysis for classification of variants of unknown clinical significance.


Asunto(s)
Discapacidad Intelectual , Trastornos del Neurodesarrollo , Metilación de ADN/genética , Genoma , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Masculino , Trastornos del Neurodesarrollo/genética , Fenotipo , Síndrome
8.
Hum Mutat ; 41(12): 2179-2194, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33131181

RESUMEN

Ciliopathies are clinically and genetically heterogeneous diseases. We studied three patients from two independent families presenting with features of Joubert syndrome: abnormal breathing pattern during infancy, developmental delay/intellectual disability, cerebellar ataxia, molar tooth sign on magnetic resonance imaging scans, and polydactyly. We identified biallelic loss-of-function (LOF) variants in CBY1, segregating with the clinical features of Joubert syndrome in the families. CBY1 localizes to the distal end of the mother centriole, contributing to the formation and function of cilia. In accordance with the clinical and mutational findings in the affected individuals, we demonstrated that depletion of Cby1 in zebrafish causes ciliopathy-related phenotypes. Levels of CBY1 transcript were found reduced in the patients compared with controls, suggesting degradation of the mutated transcript through nonsense-mediated messenger RNA decay. Accordingly, we could detect CBY1 protein in fibroblasts from controls, but not from patients by immunofluorescence. Furthermore, we observed reduced ability to ciliate, increased ciliary length, and reduced levels of the ciliary proteins AHI1 and ARL13B in patient fibroblasts. Our data show that CBY1 LOF-variants cause a ciliopathy with features of Joubert syndrome.


Asunto(s)
Anomalías Múltiples/genética , Proteínas Portadoras/genética , Cerebelo/anomalías , Ciliopatías/genética , Anomalías del Ojo/genética , Enfermedades Renales Quísticas/genética , Mutación/genética , Proteínas Nucleares/genética , Retina/anomalías , Anomalías Múltiples/diagnóstico por imagen , Anomalías Múltiples/patología , Adolescente , Animales , Cerebelo/diagnóstico por imagen , Cerebelo/patología , Niño , Preescolar , Cilios/metabolismo , Cilios/patología , Ciliopatías/diagnóstico por imagen , Ciliopatías/patología , Anomalías del Ojo/diagnóstico por imagen , Anomalías del Ojo/patología , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Homocigoto , Humanos , Lactante , Recién Nacido , Enfermedades Renales Quísticas/diagnóstico por imagen , Enfermedades Renales Quísticas/patología , Imagen por Resonancia Magnética , Masculino , Linaje , Fenotipo , Retina/diagnóstico por imagen , Retina/patología , Receptor Smoothened/metabolismo , Adulto Joven , Pez Cebra/genética
9.
Hum Mutat ; 41(10): 1738-1744, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32643838

RESUMEN

Congenital heart defects and skeletal malformations syndrome (CHDSKM) is a rare autosomal dominant disorder characterized by congenital heart disease, skeletal abnormalities, and failure to thrive. CHDSKM is caused by germline mutations in ABL1. To date, three variants have been in association with CHDSKM. In this study, we describe three de novo missense variants, c.407C>T (p.Thr136Met), c.746C>T (p.Pro249Leu), and c.1573G>A (p.Val525Met), and one recurrent variant, c.1066G>A (p.Ala356Thr), in six patients, thereby expanding the phenotypic spectrum of CHDSKM to include hearing impairment, lipodystrophy-like features, renal hypoplasia, and distinct ocular abnormalities. Functional investigation of the three novel variants showed an increased ABL1 kinase activity. The cardiac findings in additional patients with p.Ala356Thr contribute to the accumulating evidence that patients carrying either one of the recurrent variants, p.Tyr245Cys and p.Ala356Thr, have a high incidence of cardiac abnormalities. The phenotypic expansion has implications for the clinical diagnosis of CHDSKM in patients with germline ABL1 variants.


Asunto(s)
Anomalías Múltiples , Cardiopatías Congénitas , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Células Germinativas , Cardiopatías Congénitas/genética , Humanos , Fenotipo , Síndrome
10.
Hum Mol Genet ; 29(13): 2218-2239, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32504085

RESUMEN

The RNA exosome is an essential ribonuclease complex required for processing and/or degradation of both coding and non-coding RNAs. We identified five patients with biallelic variants in EXOSC5, which encodes a structural subunit of the RNA exosome. The clinical features of these patients include failure to thrive, short stature, feeding difficulties, developmental delays that affect motor skills, hypotonia and esotropia. Brain MRI revealed cerebellar hypoplasia and ventriculomegaly. While we ascertained five patients, three patients with distinct variants of EXOSC5 were studied in detail. The first patient had a deletion involving exons 5-6 of EXOSC5 and a missense variant, p.Thr114Ile, that were inherited in trans, the second patient was homozygous for p.Leu206His and the third patient had paternal isodisomy for chromosome 19 and was homozygous for p.Met148Thr. The additional two patients ascertained are siblings who had an early frameshift mutation in EXOSC5 and the p.Thr114Ile missense variant that were inherited in trans. We employed three complementary approaches to explore the requirement for EXOSC5 in brain development and assess consequences of pathogenic EXOSC5 variants. Loss of function for exosc5 in zebrafish results in shortened and curved tails/bodies, reduced eye/head size and edema. We modeled pathogenic EXOSC5 variants in both budding yeast and mammalian cells. Some of these variants cause defects in RNA exosome function as well as altered interactions with other RNA exosome subunits. These findings expand the number of genes encoding RNA exosome subunits linked to human disease while also suggesting that disease mechanism varies depending on the specific pathogenic variant.


Asunto(s)
Antígenos de Neoplasias/genética , Cerebelo/anomalías , Discapacidades del Desarrollo/genética , Enanismo/genética , Complejo Multienzimático de Ribonucleasas del Exosoma/genética , Malformaciones del Sistema Nervioso/genética , Proteínas de Unión al ARN/genética , Animales , Cerebelo/patología , Discapacidades del Desarrollo/patología , Enanismo/patología , Mutación del Sistema de Lectura/genética , Homocigoto , Humanos , Mutación Missense/genética , Malformaciones del Sistema Nervioso/patología , Linaje , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
11.
BMC Med Genet ; 21(1): 96, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32381069

RESUMEN

BACKGROUND: Joubert syndrome (JBTS) is a genetically heterogeneous group of neurodevelopmental syndromes caused by primary cilia dysfunction. Usually the neurological presentation starts with abnormal neonatal breathing followed by muscular hypotonia, psychomotor delay, and cerebellar ataxia. Cerebral MRI shows mid- and hindbrain anomalies including the molar tooth sign. We report a male patient with atypical presentation of Joubert syndrome type 23, thus expanding the phenotype. CASE PRESENTATION: Clinical features were consistent with JBTS already from infancy, yet the syndrome was not suspected before cerebral MRI later in childhood showed the characteristic molar tooth sign and ectopic neurohypophysis. From age 11 years seizures developed and after few years became increasingly difficult to treat, also related to inadequate compliance to therapy. He died at 23 years of sudden unexpected death in epilepsy (SUDEP). The genetic diagnosis remained elusive for many years, despite extensive genetic testing. We reached the genetic diagnosis by performing whole genome sequencing of the family trio and analyzing the data with the combination of one analysis pipeline for single nucleotide variants (SNVs)/indels and one for structural variants (SVs). This lead to the identification of the most common variant detected in patients with JBTS23 (OMIM# 616490), rs534542684, in compound heterozygosity with a 8.3 kb deletion in KIAA0586, not previously reported. CONCLUSIONS: We describe for the first time ectopic neurohypophysis and SUDEP in JBTS23, expanding the phenotype of this condition and raising the attention on the possible severity of the epilepsy in this disease. We also highlight the diagnostic power of WGS, which efficiently detects SNVs/indels and in addition allows the identification of SVs.


Asunto(s)
Anomalías Múltiples/genética , Proteínas de Ciclo Celular/genética , Cerebelo/anomalías , Muerte Súbita/patología , Epilepsia/genética , Anomalías del Ojo/genética , Enfermedades Renales Quísticas/genética , Retina/anomalías , Anomalías Múltiples/mortalidad , Anomalías Múltiples/patología , Adulto , Cerebelo/patología , Niño , Muerte Súbita/epidemiología , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/mortalidad , Discapacidades del Desarrollo/patología , Epilepsia/mortalidad , Epilepsia/patología , Anomalías del Ojo/mortalidad , Anomalías del Ojo/patología , Femenino , Heterocigoto , Humanos , Mutación INDEL , Enfermedades Renales Quísticas/mortalidad , Enfermedades Renales Quísticas/patología , Masculino , Neurohipófisis/metabolismo , Neurohipófisis/patología , Retina/patología , Secuenciación Completa del Genoma , Adulto Joven
13.
Cell Calcium ; 85: 102110, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31785581

RESUMEN

Calcium signaling plays a central role in bone development and homeostasis. Store operated calcium entry (SOCE) is an important calcium influx pathway mediated by calcium release activated calcium (CRAC) channels in the plasma membrane. Stromal interaction molecule 1 (STIM1) is an endoplasmic reticulum calcium sensing protein important for SOCE. We generated a mouse model expressing the STIM1 R304W mutation, causing Stormorken syndrome in humans. Stim1R304W/R304W mice showed perinatal lethality, and the only three animals that survived into adulthood presented with reduced growth, low body weight, and thoracic kyphosis. Radiographs revealed a reduced number of ribs in the Stim1R304W/R304W mice. Microcomputed tomography data revealed decreased cortical bone thickness and increased trabecular bone volume fraction in Stim1R304W/R304W mice, which had thinner and more compact bone compared to wild type mice. The Stim1R304W/+ mice showed an intermediate phenotype. Histological analyses showed that the Stim1R304W/R304W mice had abnormal bone architecture, with markedly increased number of trabeculae and reduced bone marrow cavity. Homozygous mice showed STIM1 positive osteocytes and osteoblasts. These findings highlight the critical role of the gain-of-function (GoF) STIM1 R304W protein in skeletal development and homeostasis in mice. Furthermore, the novel feature of bilateral subgingival hair growth on the lower incisors in the Stim1R304W/R304W mice and 25 % of the heterozygous mice indicate that the GoF STIM1 R304W protein also induces an abnormal epithelial cell fate.


Asunto(s)
Hueso Esponjoso/patología , Encía/crecimiento & desarrollo , Cabello/crecimiento & desarrollo , Molécula de Interacción Estromal 1/metabolismo , Animales , Huesos/anomalías , Huesos/patología , Hueso Cortical/diagnóstico por imagen , Hueso Cortical/patología , Cabello/ultraestructura , Homocigoto , Incisivo/patología , Cifosis/genética , Cifosis/patología , Megacariocitos/metabolismo , Megacariocitos/patología , Ratones , Mutación , Osteoblastos/metabolismo , Osteoblastos/patología , Osteocitos/metabolismo , Osteocitos/patología , Costillas/diagnóstico por imagen , Costillas/patología , Esplenomegalia/patología , Tórax/patología , Microtomografía por Rayos X
15.
Cell Calcium ; 76: 87-100, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30390422

RESUMEN

STIM1 and ORAI1 regulate store-operated Ca2+ entry (SOCE) in most cell types, and mutations in these proteins have deleterious and diverse effects. We established a mouse line expressing the STIM1 R304 W gain-of-function mutation causing Stormorken syndrome to explore effects on organ and cell physiology. While STIM1 R304 W was lethal in the homozygous state, surviving mice presented with reduced growth, skeletal muscle degeneration, and reduced exercise endurance. Variable STIM1 expression levels between tissues directly impacted cellular SOCE capacity. In contrast to patients with Stormorken syndrome, STIM1 was downregulated in fibroblasts from Stim1R304W/R304W mice, which maintained SOCE despite constitutive protein activity. In studies using foetal liver chimeras, STIM1 protein was undetectable in homozygous megakaryocytes and platelets, resulting in impaired platelet activation and absent SOCE. These data indicate that downregulation of STIM1 R304 W effectively opposes the gain-of-function phenotype associated with this mutation, and highlight the importance of STIM1 in skeletal muscle development and integrity.


Asunto(s)
Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Activación Plaquetaria , Molécula de Interacción Estromal 1/metabolismo , Animales , Calcio/metabolismo , Femenino , Locomoción , Masculino , Ratones , Ratones Endogámicos
16.
Thyroid ; 28(11): 1406-1415, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30296914

RESUMEN

BACKGROUND: Thyroid hormones (TH) are essential for brain development and function. The TH transporters monocarboxylate transporter 8 (MCT8) and organic anion transporter1 C1 (OATP1C1) facilitate the transport of TH across the blood-brain barrier and into glia and neuronal cells in the brain. Loss of MCT8 function causes Allan-Herndon-Dudley syndrome (AHDS, OMIM 300523) characterized by severe intellectual and motor disability due to cerebral hypothyroidism. Here, the first patient with loss of OATP1C1 function is described. The patient is a 15.5-year-old girl with normal development in the first year of life, who gradually developed dementia with spasticity and intolerance to cold. Brain imaging demonstrated gray and white matter degeneration and severe glucose hypometabolism. METHODS: Exome sequencing of the patient and parents was performed to identify the disease-causing mutation, and the effect of the mutation was studied through a panel of in vitro experiments, including thyroxine uptake studies, immunoblotting, and immunocytochemistry. Furthermore, the clinical effects of treatment with the triiodothyronine analogue triiodothyroacetic acid (Triac) are described. RESULTS: Exome sequencing identified a homozygous missense mutation in OATP1C1, changing the highly conserved aspartic acid 252 to asparagine (D252N). In vitro, the mutated OATP1C1 displays impaired plasma membrane localization and decreased cellular thyroxine uptake. After treatment with Triac, the clinical condition improved in several domains. CONCLUSIONS: This is the first report of human OATP1C1 deficiency compatible with brain-specific hypothyroidism and neurodegeneration.


Asunto(s)
Encéfalo/metabolismo , Mutación Missense , Degeneración Nerviosa/genética , Transportadores de Anión Orgánico/genética , Adolescente , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Femenino , Humanos , Degeneración Nerviosa/diagnóstico por imagen , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Transportadores de Anión Orgánico/metabolismo , Secuenciación del Exoma
17.
Nat Genet ; 50(3): 344-348, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29483653

RESUMEN

Transforming growth factor (TGF)-ß1 (encoded by TGFB1) is the prototypic member of the TGF-ß family of 33 proteins that orchestrate embryogenesis, development and tissue homeostasis1,2. Following its discovery 3 , enormous interest and numerous controversies have emerged about the role of TGF-ß in coordinating the balance of pro- and anti-oncogenic properties4,5, pro- and anti-inflammatory effects 6 , or pro- and anti-fibrinogenic characteristics 7 . Here we describe three individuals from two pedigrees with biallelic loss-of-function mutations in the TGFB1 gene who presented with severe infantile inflammatory bowel disease (IBD) and central nervous system (CNS) disease associated with epilepsy, brain atrophy and posterior leukoencephalopathy. The proteins encoded by the mutated TGFB1 alleles were characterized by impaired secretion, function or stability of the TGF-ß1-LAP complex, which is suggestive of perturbed bioavailability of TGF-ß1. Our study shows that TGF-ß1 has a critical and nonredundant role in the development and homeostasis of intestinal immunity and the CNS in humans.


Asunto(s)
Encefalopatías/complicaciones , Encefalopatías/genética , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/genética , Factor de Crecimiento Transformador beta1/genética , Análisis Mutacional de ADN , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/patología , Masculino , Linaje , Índice de Severidad de la Enfermedad
18.
Genet Med ; 20(7): 778-784, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-28837161

RESUMEN

PURPOSE: To elucidate the novel molecular cause in two unrelated consanguineous families with autosomal recessive intellectual disability. METHODS: A combination of homozygosity mapping and exome sequencing was used to locate the plausible genetic defect in family F162, while only exome sequencing was followed in the family PKMR65. The protein 3D structure was visualized with the University of California-San Francisco Chimera software. RESULTS: All five patients from both families presented with severe intellectual disability, aggressive behavior, and speech and motor delay. Four of the five patients had microcephaly. We identified homozygous missense variants in LINGO1, p.(Arg290His) in family F162 and p.(Tyr288Cys) in family PKMR65. Both variants were predicted to be pathogenic, and segregated with the phenotype in the respective families. Molecular modeling of LINGO1 suggests that both variants interfere with the glycosylation of the protein. CONCLUSION: LINGO1 is a transmembrane receptor, predominantly found in the central nervous system. Published loss-of-function studies in mouse and zebrafish have established a crucial role of LINGO1 in normal neuronal development and central nervous system myelination by negatively regulating oligodendrocyte differentiation and neuronal survival. Taken together, our results indicate that biallelic LINGO1 missense variants cause autosomal recessive intellectual disability in humans.


Asunto(s)
Discapacidad Intelectual/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Alelos , Mapeo Cromosómico/métodos , Familia , Femenino , Frecuencia de los Genes/genética , Genotipo , Homocigoto , Humanos , Trastornos del Desarrollo del Lenguaje/genética , Masculino , Proteínas de la Membrana/fisiología , Microcefalia/genética , Actividad Motora/genética , Mutación Missense/genética , Proteínas del Tejido Nervioso/fisiología , Pakistán , Linaje , Fenotipo , Análisis de Secuencia de Proteína , Secuenciación del Exoma
19.
Am J Hum Genet ; 100(6): 907-925, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28575647

RESUMEN

Yin and yang 1 (YY1) is a well-known zinc-finger transcription factor with crucial roles in normal development and malignancy. YY1 acts both as a repressor and as an activator of gene expression. We have identified 23 individuals with de novo mutations or deletions of YY1 and phenotypic features that define a syndrome of cognitive impairment, behavioral alterations, intrauterine growth restriction, feeding problems, and various congenital malformations. Our combined clinical and molecular data define "YY1 syndrome" as a haploinsufficiency syndrome. Through immunoprecipitation of YY1-bound chromatin from affected individuals' cells with antibodies recognizing both ends of the protein, we show that YY1 deletions and missense mutations lead to a global loss of YY1 binding with a preferential retention at high-occupancy sites. Finally, we uncover a widespread loss of H3K27 acetylation in particular on the YY1-bound enhancers, underscoring a crucial role for YY1 in enhancer regulation. Collectively, these results define a clinical syndrome caused by haploinsufficiency of YY1 through dysregulation of key transcriptional regulators.


Asunto(s)
Cromatina/metabolismo , Haploinsuficiencia/genética , Discapacidad Intelectual/genética , Transcripción Genética , Factor de Transcripción YY1/genética , Acetilación , Adolescente , Secuencia de Bases , Preescolar , Inmunoprecipitación de Cromatina , Estudios de Cohortes , Elementos de Facilitación Genéticos/genética , Femenino , Ontología de Genes , Haplotipos/genética , Hemicigoto , Histonas/metabolismo , Humanos , Linfocitos/metabolismo , Masculino , Metilación , Modelos Moleculares , Mutación Missense/genética , Unión Proteica/genética , Dominios Proteicos , Factor de Transcripción YY1/química
20.
Genes (Basel) ; 7(12)2016 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-27916860

RESUMEN

Biallelic PIGT variants were previously reported in seven patients from three families with Multiple Congenital Anomalies-Hypotonia Seizures Syndrome 3 (MCAHS3), characterized by epileptic encephalopathy, hypotonia, global developmental delay/intellectual disability, cerebral and cerebellar atrophy, craniofacial dysmorphisms, and skeletal, ophthalmological, cardiac, and genitourinary abnormalities. We report a novel homozygous PIGT missense variant c.1079G>T (p.Gly360Val) in two brothers with several of the typical features of MCAHS3, but in addition, pyramidal tract neurological signs. Notably, they are the first patients with MCAHS3 without skeletal, cardiac, or genitourinary anomalies. PIGT encodes a crucial subunit of the glycosylphosphatidylinositol (GPI) transamidase complex, which catalyzes the attachment of proteins to GPI-anchors, attaching the proteins to the cell membrane. In vitro studies in cells from the two brothers showed reduced levels of GPI-anchors and GPI-anchored proteins on the cell surface, supporting the pathogenicity of the novel PIGT variant.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA