Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Allergy Clin Immunol ; 149(3): 1120-1127.e8, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34416217

RESUMEN

BACKGROUND: Proteasome-associated autoinflammatory syndromes (PRAASs) form a family of recently described rare autosomal recessive disorders of disturbed proteasome assembly and proteolytic activity caused by mutations in genes coding for proteasome subunits. The treatment options for these proteasome disorders consist of lifelong immunosuppressive drugs or Janus kinase inhibitors, which may have partial efficacy and noticeable side effects. Because proteasomes are ubiquitously expressed, it is unknown whether hematopoietic stem cell transplantation (HSCT) may be a sufficient treatment option. OBJECTIVE: Our aim was to report the case of a young boy with a treatment-resistant cutaneous vasculitis that was initially suspected to be associated with a gene variant in SH2D1A. METHODS: Whole-exome sequencing was performed to identify the genetic defect. Molecular and functional analyses were performed to assess the impact of variants on proteasomal function. The immune characterization led to the decision to perform HSCT on our patient and conduct follow-up over the 7-year period after the transplant. Because loss of myeloid chimerism after the first HSCT was associated with relapse of autoinflammation, a second HSCT was performed. RESULTS: After the successful second HSCT, the patient developed mild symptoms of lipodystrophy, which raised the suspicion of a PRAAS. Genetic analysis revealed 2 novel heterozygous variants in PSMB4 (encoding proteasomal subunit ß7). Retrospective analysis of patient cells stored before the first HSCT and patient cells obtained after the second HSCT demonstrated that HSCT successfully rescued proteasome function, restored protein homeostasis, and resolved the interferon-stimulated gene signature. Furthermore, successful HSCT alleviated the autoinflammatory manifestations in our patient. CONCLUSION: Patients with treatment-resistant PRAAS can be cured by HSCT.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Lipodistrofia , Niño , Humanos , Lipodistrofia/genética , Masculino , Complejo de la Endopetidasa Proteasomal/genética , Estudios Retrospectivos , Síndrome
2.
J Psychiatr Res ; 138: 125-129, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33848968

RESUMEN

Inborn errors of metabolism (IEMs) are a group of rare genetic disorders which, when emerging later in life, are often characterized by neuropsychiatric manifestations including psychosis. This study aimed to determine whether it would be useful to screen patients presenting with a psychotic disorder for IEMs by a single blood sample using Next Generation Sequencing (NGS), in order to detect rare, treatable causes of psychotic disorders. Blood was drawn from 60 patients with a psychotic disorder, with a duration of illness of less than 5 years. Blood samples were screened for 67 genes using NGS (Illumina® MiSeq sequencing technique). The results were compared to the human reference genome (GoNL, n = 498). The identified variants were classified according to the ACMG classification. For the psychotic patients, 6 variants of a likely pathogenic (class 4, n = 2) or pathogenic (class 5, n = 4) origin were found. As all variants were heterozygous, no patients were considered to be affected by an IEM. For the GoNL control group, 73 variants of a likely pathogenic (class 4, n = 31) or pathogenic (class 5, n = 42) origin were found. All of these found variants were heterozygous. Therefore, these individuals from the control group were considered to be a carrier only. Thus, no patients were identified to have an IEM as an underlying disease using this approach. However, NGS may be useful to detect variants of genes associated with IEMs in an enriched subgroup of psychotic patients.


Asunto(s)
Trastornos Mentales , Errores Innatos del Metabolismo , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Tamizaje Masivo , Errores Innatos del Metabolismo/diagnóstico , Errores Innatos del Metabolismo/genética
3.
J Neuromuscul Dis ; 8(2): 261-272, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33386810

RESUMEN

BACKGROUND: A Dutch cohort of 105 carefully selected limb girdle muscular dystrophy (LGMD) patients from 68 families has been subject to genetic testing over the last 20 years. After subsequent targeted gene analysis around two thirds (45/68) of the families had received a genetic diagnosis in 2013. OBJECTIVE: To describe the results of further genetic testing in the remaining undiagnosed limb girdle muscular dystrophy families in this cohort. METHODS: In the families of the cohort for whom no genetic diagnosis was established (n = 23) further testing using Sanger sequencing, next generation sequencing with gene panel analysis or whole-exome sequencing was performed. In one case DNA analysis for facioscapulohumeral dystrophy type 1 was carried out. RESULTS: In eight families no additional genetic tests could be performed. In 12 of the remaining 15 families in which additional testing could be performed a genetic diagnosis was established: two LGMDR1 calpain3-related families with CAPN3 mutations, one LGMDR2 dysferlin-related family with DYSF mutations, three sarcoglycanopathy families (LGMDR3-5 α-, ß- and γ-sarcoglycan-related) with SGCA/SGCB/SGCG mutations, one LGMDR8 TRIM 32-related family with TRIM32 mutations, two LGMDR19 GMPPB-related families with GMPPB mutations, one family with MICU1-related myopathy, one family with FLNC-related myopathy and one family with facioscapulohumeral dystrophy type 1. At this moment a genetic diagnosis has been made in 57 of the 60 families of which DNA was available (95%). CONCLUSION: A genetic diagnosis is obtained in 95% of the families of the original Dutch LGMD cohort of which DNA was available.


Asunto(s)
Distrofia Muscular de Cinturas/genética , Adolescente , Adulto , Proteínas de Unión al Calcio , Calpaína , Proteínas de Transporte de Catión , Niño , Disferlina , Femenino , Pruebas Genéticas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Transporte de Membrana Mitocondrial , Proteínas Musculares , Distrofias Musculares/genética , Mutación , Países Bajos , Fenotipo , Sarcoglicanopatías/genética , Análisis de Secuencia de ADN , Factores de Transcripción , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Secuenciación del Exoma , Adulto Joven
4.
J Med Genet ; 57(12): 808-819, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32409512

RESUMEN

INTRODUCTION: Pigmentary mosaicism (PM) manifests by pigmentation anomalies along Blaschko's lines and represents a clue toward the molecular diagnosis of syndromic intellectual disability (ID). Together with new insights on the role for lysosomal signalling in embryonic stem cell differentiation, mutations in the X-linked transcription factor 3 (TFE3) have recently been reported in five patients. Functional analysis suggested these mutations to result in ectopic nuclear gain of functions. MATERIALS AND METHODS: Subsequent data sharing allowed the clustering of de novo TFE3 variants identified by exome sequencing on DNA extracted from leucocytes in patients referred for syndromic ID with or without PM. RESULTS: We describe the detailed clinical and molecular data of 17 individuals harbouring a de novo TFE3 variant, including the patients that initially allowed reporting TFE3 as a new disease-causing gene. The 12 females and 5 males presented with pigmentation anomalies on Blaschko's lines, severe ID, epilepsy, storage disorder-like features, growth retardation and recognisable facial dysmorphism. The variant was at a mosaic state in at least two male patients. All variants were missense except one splice variant. Eleven of the 13 variants were localised in exon 4, 2 in exon 3, and 3 were recurrent variants. CONCLUSION: This series further delineates the specific storage disorder-like phenotype with PM ascribed to de novo TFE3 mutation in exons 3 and 4. It confirms the identification of a novel X-linked human condition associated with mosaicism and dysregulation within the mechanistic target of rapamycin (mTOR) pathway, as well as a link between lysosomal signalling and human development.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Epilepsia/genética , Discapacidad Intelectual/genética , Trastornos de la Pigmentación/genética , Adolescente , Adulto , Niño , Preescolar , Epilepsia/complicaciones , Epilepsia/patología , Femenino , Genes Ligados a X/genética , Humanos , Lactante , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/patología , Masculino , Mosaicismo , Patología Molecular/normas , Trastornos de la Pigmentación/complicaciones , Trastornos de la Pigmentación/patología , Secuenciación del Exoma , Adulto Joven
5.
Pediatr Neurol ; 102: 62-66, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31371121

RESUMEN

BACKGROUND: Hereditary folate malabsorption is a multisystem disease owing to biallelic variants in the gene encoding the proton-coupled folate transporter. Hereditary folate malabsorption is treated with folinic acid, aimed to restore blood and cerebrospinal fluid folate levels. Little is known as to whether oral or intramuscular supplementation of folinic acid is most effective. METHODS: Here we describe a one-year-old boy with hereditary folate malabsorption presenting with the typical features including failure to thrive, aphthous stomatitis, macrocytic anemia along with severe developmental impairment and epilepsy, as well as a magnetic resonance imaging of the brain showing bilateral occipital, cortical calcifications characteristic of hereditary folate malabsorption. We compared the effect of treatment with oral folinic acid versus intramuscular folinic acid supplementation by measuring plasma and cerebrospinal fluid folate levels. RESULTS: Compared with oral administration, intramuscular treatment resulted in higher folate levels in blood and, most importantly, normalization of folate levels in cerebrospinal fluid. Clinically, nearly all systemic and neurological symptoms resolved. CONCLUSION: Normal cerebrospinal fluid folate levels can be achieved in individuals with hereditary folate malabsorption with intramuscular (but not with oral) administration of folinic acid.


Asunto(s)
Deficiencia de Ácido Fólico/tratamiento farmacológico , Leucovorina/farmacología , Síndromes de Malabsorción/tratamiento farmacológico , Complejo Vitamínico B/farmacología , Humanos , Lactante , Inyecciones Intramusculares , Leucovorina/administración & dosificación , Masculino , Complejo Vitamínico B/administración & dosificación
6.
Am J Hum Genet ; 104(6): 1040-1059, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31079900

RESUMEN

The heterogeneous nuclear ribonucleoprotein (HNRNP) genes code for a set of RNA-binding proteins that function primarily in the spliceosome C complex. Pathogenic variants in these genes can drive neurodegeneration, through a mechanism involving excessive stress-granule formation, or developmental defects, through mechanisms that are not known. Here, we report four unrelated individuals who have truncating or missense variants in the same C-terminal region of hnRNPR and who have multisystem developmental defects including abnormalities of the brain and skeleton, dysmorphic facies, brachydactyly, seizures, and hypoplastic external genitalia. We further identified in the literature a fifth individual with a truncating variant. RNA sequencing of primary fibroblasts reveals that these HNRNPR variants drive significant changes in the expression of several homeobox genes, as well as other transcription factors, such as LHX9, TBX1, and multiple HOX genes, that are considered fundamental regulators of embryonic and gonad development. Higher levels of retained intronic HOX sequences and lost splicing events in the HOX cluster are observed in cells carrying HNRNPR variants, suggesting that impaired splicing is at least partially driving HOX deregulation. At basal levels, stress-granule formation appears normal in primary and transfected cells expressing HNRNPR variants. However, these cells reveal profound recovery defects, where stress granules fail to disassemble properly, after exposure to oxidative stress. This study establishes an essential role for HNRNPR in human development and points to a mechanism that may unify other "spliceosomopathies" linked to variants that drive multi-system congenital defects and are found in hnRNPs.


Asunto(s)
Discapacidades del Desarrollo/etiología , Fibroblastos/patología , Regulación de la Expresión Génica , Genes Homeobox/genética , Ribonucleoproteínas Nucleares Heterogéneas/genética , Mutación , Empalme del ARN/genética , Niño , Preescolar , Discapacidades del Desarrollo/patología , Femenino , Fibroblastos/metabolismo , Humanos , Lactante , Masculino , Estrés Oxidativo , Fenotipo , Secuenciación del Exoma
7.
Genet Med ; 21(10): 2355-2363, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30940925

RESUMEN

PURPOSE: A new syndrome with hypotonia, intellectual disability, and eye abnormalities (HIDEA) was previously described in a large consanguineous family. Linkage analysis identified the recessive disease locus, and genome sequencing yielded three candidate genes with potentially pathogenic biallelic variants: transketolase (TKT), transmembrane prolyl 4-hydroxylase (P4HTM), and ubiquitin specific peptidase 4 (USP4). However, the causative gene remained elusive. METHODS: International collaboration and exome sequencing were used to identify new patients with HIDEA and biallelic, potentially pathogenic, P4HTM variants. Segregation analysis was performed using Sanger sequencing. P4H-TM wild-type and variant constructs without the transmembrane region were overexpressed in insect cells and analyzed using sodium dodecyl sulfate-polyacrylamide gel electrophoresis and western blot. RESULTS: Five different homozygous or compound heterozygous pathogenic P4HTM gene variants were identified in six new and six previously published patients presenting with HIDEA. Hypoventilation, obstructive and central sleep apnea, and dysautonomia were identified as novel features associated with the phenotype. Characterization of three of the P4H-TM variants demonstrated yielding insoluble protein products and, thus, loss-of-function. CONCLUSIONS: Biallelic loss-of-function P4HTM variants were shown to cause HIDEA syndrome. Our findings enable diagnosis of the condition, and highlight the importance of assessing the need for noninvasive ventilatory support in patients.


Asunto(s)
Prolil Hidroxilasas/genética , Transcetolasa/genética , Proteasas Ubiquitina-Específicas/genética , Anomalías Múltiples/genética , Adolescente , Adulto , Niño , Preescolar , Epilepsia/genética , Exoma , Anomalías del Ojo/genética , Femenino , Humanos , Hipoventilación/genética , Discapacidad Intelectual/genética , Mutación con Pérdida de Función/genética , Masculino , Hipotonía Muscular/genética , Linaje , Fenotipo , Disautonomías Primarias/genética , Prolil Hidroxilasas/metabolismo , Síndrome , Transcetolasa/metabolismo , Secuenciación del Exoma , Adulto Joven
8.
Cell Stem Cell ; 24(2): 257-270.e8, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30595499

RESUMEN

Self-renewal and differentiation of pluripotent murine embryonic stem cells (ESCs) is regulated by extrinsic signaling pathways. It is less clear whether cellular metabolism instructs developmental progression. In an unbiased genome-wide CRISPR/Cas9 screen, we identified components of a conserved amino-acid-sensing pathway as critical drivers of ESC differentiation. Functional analysis revealed that lysosome activity, the Ragulator protein complex, and the tumor-suppressor protein Folliculin enable the Rag GTPases C and D to bind and seclude the bHLH transcription factor Tfe3 in the cytoplasm. In contrast, ectopic nuclear Tfe3 represses specific developmental and metabolic transcriptional programs that are associated with peri-implantation development. We show differentiation-specific and non-canonical regulation of Rag GTPase in ESCs and, importantly, identify point mutations in a Tfe3 domain required for cytoplasmic inactivation as potentially causal for a human developmental disorder. Our work reveals an instructive and biomedically relevant role of metabolic signaling in licensing embryonic cell fate transitions.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Diferenciación Celular , Lisosomas/metabolismo , Transducción de Señal , Alelos , Animales , Autorrenovación de las Células , Femenino , GTP Fosfohidrolasas/metabolismo , Genoma , Humanos , Masculino , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Fosforilación , Mutación Puntual/genética , Unión Proteica , Transcripción Genética
9.
Genes Dev ; 32(5-6): 373-388, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29555651

RESUMEN

It has been well established that histone and DNA modifications are critical to maintaining the equilibrium between pluripotency and differentiation during early embryogenesis. Mutations in key regulators of DNA methylation have shown that the balance between gene regulation and function is critical during neural development in early years of life. However, there have been no identified cases linking epigenetic regulators to aberrant human development and fetal demise. Here, we demonstrate that a homozygous inactivating mutation in the histone deacetylase SIRT6 results in severe congenital anomalies and perinatal lethality in four affected fetuses. In vitro, the amino acid change at Asp63 to a histidine results in virtually complete loss of H3K9 deacetylase and demyristoylase functions. Functionally, SIRT6 D63H mouse embryonic stem cells (mESCs) fail to repress pluripotent gene expression, direct targets of SIRT6, and exhibit an even more severe phenotype than Sirt6-deficient ESCs when differentiated into embryoid bodies (EBs). When terminally differentiated toward cardiomyocyte lineage, D63H mutant mESCs maintain expression of pluripotent genes and fail to form functional cardiomyocyte foci. Last, human induced pluripotent stem cells (iPSCs) derived from D63H homozygous fetuses fail to differentiate into EBs, functional cardiomyocytes, and neural progenitor cells due to a failure to repress pluripotent genes. Altogether, our study described a germline mutation in SIRT6 as a cause for fetal demise, defining SIRT6 as a key factor in human development and identifying the first mutation in a chromatin factor behind a human syndrome of perinatal lethality.


Asunto(s)
Mutación/genética , Sirtuinas/genética , Animales , Diferenciación Celular/genética , Cuerpos Embrioides , Células Madre Embrionarias , Muerte Fetal , Expresión Génica/genética , Humanos , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
10.
J Inherit Metab Dis ; 41(5): 897-898, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29464431

RESUMEN

Mutations in the SEPSECS gene are associated with pontocerebellar hypoplasia type 2D. Pontocerebellar hypoplasia (PCH) is a heterogeneous group of rare autosomal recessive neurodegenerative disorders, mainly affecting pons and cerebellum. Patients have severe motor and cognitive impairments and often die during infancy. Here, we report a 23-year-old woman with slowly progressive cerebellar ataxia and cognitive impairment, in whom a homozygous missense mutation in the SEPSECS gene (c.1321G>A; p.Gly441Arg) was identified with whole exome sequencing. Our findings underline that defects in selenoprotein synthesis can also result in milder cerebellar atrophy presenting at a later age.


Asunto(s)
Aminoacil-ARNt Sintetasas/genética , Encéfalo/patología , Ataxia Cerebelosa/etiología , Enfermedades Cerebelosas/fisiopatología , Microcefalia/etiología , Encéfalo/diagnóstico por imagen , Enfermedades Cerebelosas/genética , Disfunción Cognitiva/etiología , Femenino , Homocigoto , Humanos , Imagen por Resonancia Magnética , Mutación Missense , Secuenciación del Exoma , Adulto Joven
11.
Eur J Med Genet ; 61(2): 94-97, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29056561

RESUMEN

Bloom syndrome is an autosomal recessive condition characterized by severe pre- and postnatal growth deficiency, immunodeficiency, an increased risk for malignancies, craniofacial dysmorphisms, and "typical" erythematous sun-sensitive skin lesions of the face. This facial rash has a butterfly-shaped distribution around the nose and is usually observed for the first time during the early years of life. Though reported as being a main feature of Bloom syndrome, there seems to be phenotypic variability regarding this facial skin rash among patients. It has been previously reported that in some individuals with Bloom syndrome these sun-sensitive lesions are less prominent or even absent. In this report we describe a 36 year old woman with short stature, microcephaly, several dysmorphisms, congenital hypothyroidism and premature ovarian failure. She was diagnosed with nasopharyngeal carcinoma at 36 years of age, only a few months after her consultation at the department of Clinical Genetics. Whole Exome Sequencing demonstrated that she had Bloom syndrome caused by a compound heterozygous mutation in BLM (c.2207_2212delinsTAGATTC; p.(Tyr736Leufs*5) and c.3681del; p.(Lys1227Asnfs*52)). She did not have facial sun-sensitive erythematous rash during childhood nor adulthood. We conclude that Bloom syndrome does not always present with erythematous sun-sensitive skin lesions of the face. We would like to underline that phenotypic variation regarding this "hallmark" feature of Bloom syndrome exists. Being aware of this might prevent a delay in diagnosing this rare short-stature syndrome and, subsequently, its potential clinical implications.


Asunto(s)
Síndrome de Bloom/patología , Eritema/patología , Fenotipo , Adulto , Síndrome de Bloom/genética , Diagnóstico Diferencial , Eritema/etiología , Eritema/genética , Femenino , Humanos , RecQ Helicasas/genética , Luz Solar/efectos adversos
12.
Am J Med Genet A ; 170A(2): 510-514, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26601923

RESUMEN

We report on a boy with a neonatal short limb skeletal dysplasia with serious medical complications, associated with one intragenic and one complete deletion of XYLT1. XYLT1 mutations have recently been reported as causative in recessive Desbuquois skeletal dysplasia (DBSD), but the skeletal features in our patient do not fit this diagnosis. It is possible that the phenotype of XYLT1 mutations extends to more aspecific types of short limb skeletal dysplasias and not to DBSD alone.


Asunto(s)
Extremidades/embriología , Anomalías Musculoesqueléticas/genética , Osteocondrodisplasias/genética , Pentosiltransferasa/genética , Eliminación de Secuencia/genética , Extremidades/patología , Humanos , Recién Nacido , Masculino , Anomalías Musculoesqueléticas/diagnóstico , Osteocondrodisplasias/diagnóstico , Fenotipo , UDP Xilosa Proteína Xilosiltransferasa
13.
J Mol Biol ; 411(2): 397-416, 2011 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-21699900

RESUMEN

FSHD region gene 1 (FRG1) is a dynamic nuclear and cytoplasmic protein that, in skeletal muscle, shows additional localization to the sarcomere. Maintaining appropriate levels of FRG1 protein is critical for muscular and vascular development in vertebrates; however, its precise molecular function is unknown. This study investigates the molecular functions of human FRG1, along with mouse FRG1 and Xenopus frg1, using molecular, biochemical, and cellular-biological approaches, to provide further insight into its roles in vertebrate development. The nuclear fraction of the endogenous FRG1 is localized in nucleoli, Cajal bodies, and actively transcribed chromatin; however, contrary to overexpressed FRG1, the endogenous FRG1 is not associated with nuclear speckles. We characterize the nuclear and nucleolar import of FRG1, the potential effect of phosphorylation, and its interaction with the importin karyopherin α2. Consistent with a role in RNA biogenesis, human FRG1 is associated with mRNA in vivo and invitro, interacts directly with TAP (Tip-associated protein; the major mRNA export receptor), and is a dynamic nuclear-cytoplasmic shuttling protein supporting a function for FRG1 in mRNA transport. Biochemically, we characterize FRG1 actin binding activity and show that the cytoplasmic pool of FRG1 is dependent on an intact actin cytoskeleton for its localization. These data provide the first biochemical activities (actin binding and RNA binding) for human FRG1 and the characterization of the endogenous human FRG1, together indicating that FRG1 is involved in multiple aspects of RNA biogenesis, including mRNA transport and, potentially, cytoplasmic mRNA localization.


Asunto(s)
Actinas/metabolismo , Proteínas Nucleares/metabolismo , ARN Mensajero/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Nucléolo Celular/química , Núcleo Celular/química , Citoplasma/química , Humanos , Carioferinas/metabolismo , Ratones , Proteínas de Microfilamentos , Modelos Biológicos , Unión Proteica , Proteínas/metabolismo , Proteínas de Unión al ARN , Xenopus , Proteínas de Xenopus/metabolismo
14.
Mol Biol Cell ; 21(21): 3735-48, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20826608

RESUMEN

The nuclear space is mostly occupied by chromosome territories and nuclear bodies. Although this organization of chromosomes affects gene function, relatively little is known about the role of nuclear bodies in the organization of chromosomal regions. The nucleolus is the best-studied subnuclear structure and forms around the rRNA repeat gene clusters on the acrocentric chromosomes. In addition to rDNA, other chromatin sequences also surround the nucleolar surface and may even loop into the nucleolus. These additional nucleolar-associated domains (NADs) have not been well characterized. We present here a whole-genome, high-resolution analysis of chromatin endogenously associated with nucleoli. We have used a combination of three complementary approaches, namely fluorescence comparative genome hybridization, high-throughput deep DNA sequencing and photoactivation combined with time-lapse fluorescence microscopy. The data show that specific sequences from most human chromosomes, in addition to the rDNA repeat units, associate with nucleoli in a reproducible and heritable manner. NADs have in common a high density of AT-rich sequence elements, low gene density and a statistically significant enrichment in transcriptionally repressed genes. Unexpectedly, both the direct DNA sequencing and fluorescence photoactivation data show that certain chromatin loci can specifically associate with either the nucleolus, or the nuclear envelope.


Asunto(s)
Nucléolo Celular/diagnóstico por imagen , Nucléolo Celular/genética , Cromatina/genética , Cromosomas Humanos/genética , Ciclo Celular , Línea Celular Tumoral , Hibridación Genómica Comparativa , Biología Computacional , ADN Ribosómico/metabolismo , Fluorescencia , Genes de ARNr , Estudio de Asociación del Genoma Completo/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hibridación in Situ , Microscopía Fluorescente , Datos de Secuencia Molecular , Análisis de la Célula Individual , Imagen de Lapso de Tiempo , Ultrasonografía
15.
Mol Biol Cell ; 21(9): 1569-84, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20219969

RESUMEN

Human small nucleolar RNAs (snoRNAs) that copurify with nucleoli isolated from HeLa cells have been characterized. Novel fibrillarin-associated snoRNAs were detected that allowed the creation of a new vector system for the targeted knockdown of one or more genes in mammalian cells. The snoMEN (snoRNA modulator of gene expressioN) vector technology is based on snoRNA HBII-180C, which contains an internal sequence that can be manipulated to make it complementary to RNA targets. Gene-specific knockdowns are demonstrated for endogenous cellular proteins and for G/YFP-fusion proteins. Multiplex snoMEN vectors coexpress multiple snoRNAs in one transcript, targeted either to different genes or to different sites in the same gene. Protein replacement snoMEN vectors can express a single transcript combining cDNA for a tagged protein with introns containing cognate snoRNAs targeted to knockdown the endogenous cellular protein. We foresee applications for snoMEN vectors in basic gene expression research, target validation, and gene therapy.


Asunto(s)
Expresión Génica , Técnicas de Silenciamiento del Gen/métodos , Vectores Genéticos/genética , ARN Nucleolar Pequeño/genética , Nucléolo Celular/genética , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Clonación Molecular , Biblioteca de Genes , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Hibridación Fluorescente in Situ , Microscopía Fluorescente , Mutación , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Transfección
16.
Nucleus ; 1(3): 284-97, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21327076

RESUMEN

This live cell study of chromatin dynamics in four dimensions (space and time) in cycling human cells provides direct evidence for three hypotheses first proposed by Theodor Boveri in seminal studies of fixed blastomeres from Parascaris equorum embryos: (I) Chromosome territory (CT) arrangements are stably maintained during interphase. (II) Chromosome proximity patterns change profoundly during prometaphase. (III) Similar CT proximity patterns in pairs of daughter nuclei reflect symmetrical chromosomal movements during anaphase and telophase, but differ substantially from the arrangement in mother cell nucleus. Hypothesis I could be confirmed for the majority of interphase cells. A minority, however, showed complex, rotational movements of CT assemblies with large-scale changes of CT proximity patterns, while radial nuclear arrangements were maintained. A new model of chromatin dynamics is proposed. It suggests that long-range DNA-DNA interactions in cell nuclei may depend on a combination of rotational CT movements and locally constrained chromatin movements.


Asunto(s)
Ciclo Celular , Cromatina/metabolismo , Animales , Línea Celular , Núcleo Celular/metabolismo , Posicionamiento de Cromosoma , Cromosomas Humanos/metabolismo , Humanos , Modelos Biológicos , Movimiento , Reproducibilidad de los Resultados , Factores de Tiempo
17.
Nat Rev Mol Cell Biol ; 8(7): 574-85, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17519961

RESUMEN

The nucleolus is a distinct subnuclear compartment that was first observed more than 200 years ago. Nucleoli assemble around the tandemly repeated ribosomal DNA gene clusters and 28S, 18S and 5.8S ribosomal RNAs (rRNAs) are transcribed as a single precursor, which is processed and assembled with the 5S rRNA into ribosome subunits. Although the nucleolus is primarily associated with ribosome biogenesis, several lines of evidence now show that it has additional functions. Some of these functions, such as regulation of mitosis, cell-cycle progression and proliferation, many forms of stress response and biogenesis of multiple ribonucleoprotein particles, will be discussed, as will the relation of the nucleolus to human diseases.


Asunto(s)
Nucléolo Celular/metabolismo , Animales , Nucléolo Celular/química , Nucléolo Celular/genética , Nucléolo Celular/fisiología , Nucléolo Celular/ultraestructura , ADN Ribosómico/análisis , ADN Ribosómico/biosíntesis , Colorantes Fluorescentes , Humanos , Indoles , Microscopía Fluorescente , Mitosis , Modelos Biológicos , Región Organizadora del Nucléolo/fisiología , Región Organizadora del Nucléolo/ultraestructura , Precursores del ARN/metabolismo , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , ARN Ribosómico 18S/genética , ARN Ribosómico 18S/metabolismo , ARN Ribosómico 28S/genética , ARN Ribosómico 28S/metabolismo , ARN Ribosómico 5.8S/genética , ARN Ribosómico 5.8S/metabolismo , ARN Ribosómico 5S/biosíntesis , ARN Ribosómico 5S/genética , ARN Nucleolar Pequeño/genética , ARN Nucleolar Pequeño/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Ribosomas/genética , Ribosomas/metabolismo
18.
Chromosoma ; 116(1): 53-64, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17103222

RESUMEN

FRG1 is considered a candidate gene for facioscapulohumeral muscular dystrophy (FSHD) based on its location at chromosome 4qter and its upregulation in FSHD muscle. The FRG1 protein (FRG1P) localizes to nucleoli, Cajal bodies (and speckles), and has been suggested to be a component of the human spliceosome but its exact function is unknown. Recently, transgenic mice overexpressing high levels of FRG1P in skeletal muscle were described to present with muscular dystrophy. Moreover, upregulation of FRG1P was demonstrated to correlate with missplicing of specific pre-mRNAs. In this study, we have combined colocalization studies with yeast two-hybrid screens to identify proteins that associate with FRG1P. We demonstrate that artificially induced nucleolar aggregates of VSV-FRG1P specifically sequester proteins involved in pre-mRNA processing. In addition, we have identified SMN, PABPN1, and FAM71B, a novel speckle and Cajal body protein, as binding partners of FRG1P. All these proteins are, or seem to be, involved in RNA biogenesis. Our data confirm the presence of FRG1P in protein complexes containing human spliceosomes and support a potential role of FRG1P in either splicing or another step in nuclear RNA biogenesis. Intriguingly, among FRG1P-associated proteins are SMN and PABPN1, both being involved in neuromuscular disorders, possibly through RNA biogenesis-related processes.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas/metabolismo , Precursores del ARN/metabolismo , Procesamiento Postranscripcional del ARN , Empalme Alternativo , Animales , Línea Celular , Nucléolo Celular/metabolismo , Humanos , Inmunoprecipitación , Proteínas de Microfilamentos , Distrofia Muscular Facioescapulohumeral/genética , Proteínas Nucleares/genética , Proteínas/genética , Precursores del ARN/genética , Proteínas de Unión al ARN , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Troponina T/genética , Troponina T/metabolismo , Técnicas del Sistema de Dos Híbridos
19.
Hum Mol Genet ; 15(1): 105-11, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16319127

RESUMEN

Oculopharyngeal muscular dystrophy (OPMD) belongs to the group of protein aggregation disorders and is caused by extensions of the N-terminal polyalanine stretch of the nuclear polyA-binding protein 1 (PABPN1). The presence of PABPN1-containing intranuclear aggregates in skeletal muscle is unique for OPMD and is also observed in transgenic mouse and cell models for OPMD. These models consistently support a direct role for the protein aggregation in OPMD pathogenesis. We have isolated and characterized a diverse panel of single-domain antibody reagents (VHH), recognizing different epitopes in PABPN1. The antibody reagents specifically detect endogenous PABPN1 in cell lysates on western blot and label PABPN1 in cultured cells and muscle sections. When expressed intracellularly as intrabodies in a cellular model for OPMD, aggregation of PABPN1 was prevented in a dose-dependent manner. More importantly yet, these intrabodies could also reduce the presence of already existing aggregates. Given the domain specificity of VHH-mediated aggregation interference, this approach at least allows the definition of the nucleation kernel in aggregation-prone proteins, thus facilitating etiological insight into this and other protein aggregation disorders, and ultimately, it may well provide useful therapeutic agents.


Asunto(s)
Anticuerpos/metabolismo , Cuerpos de Inclusión/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Oculofaríngea/tratamiento farmacológico , Proteína II de Unión a Poli(A)/genética , Animales , Anticuerpos/uso terapéutico , Western Blotting , Células COS , Chlorocebus aethiops , Relación Dosis-Respuesta a Droga , Mapeo Epitopo , Células HeLa , Humanos , Distrofia Muscular Oculofaríngea/genética , Distrofia Muscular Oculofaríngea/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...